1
|
Puarattana-aroonkorn S, Tharakaraman K, Suriyawipada D, Ruchirawat M, Fuangthong M, Sasisekharan R, Artpradit C. Rapid and Scalable Production of Functional SARS-CoV-2 Virus-like Particles (VLPs) by a Stable HEK293 Cell Pool. Vaccines (Basel) 2024; 12:561. [PMID: 38932290 PMCID: PMC11209123 DOI: 10.3390/vaccines12060561] [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: 03/30/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/28/2024] Open
Abstract
At times of pandemics, such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the situation demands rapid development and production timelines of safe and effective vaccines for delivering life-saving medications quickly to patients. Typical biologics production relies on using the lengthy and arduous approach of stable single-cell clones. Here, we used an alternative approach, a stable cell pool that takes only weeks to generate compared to a stable single-cell clone that needs several months to complete. We employed the membrane, envelope, and highly immunogenic spike proteins of SARS-CoV-2 to produce virus-like particles (VLPs) using the HEK293-F cell line as a host system with an economical transfection reagent. The cell pool showed the stability of protein expression for more than one month. We demonstrated that the production of SARS-CoV-2 VLPs using this cell pool was scalable up to a stirred-tank 2 L bioreactor in fed-batch mode. The purified VLPs were properly assembled, and their size was consistent with the authentic virus. Our particles were functional as they specifically entered the cell that naturally expresses ACE-2. Notably, this work reports a practical and cost-effective manufacturing platform for scalable SARS-CoV-2 VLPs production and chromatographic purification.
Collapse
Affiliation(s)
| | - Kannan Tharakaraman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Disapan Suriyawipada
- Translational Research Unit, Chulabhorn Research Institute, Bangkok 10210, Thailand (M.F.)
| | - Mathuros Ruchirawat
- Translational Research Unit, Chulabhorn Research Institute, Bangkok 10210, Thailand (M.F.)
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok 10400, Thailand
| | - Mayuree Fuangthong
- Translational Research Unit, Chulabhorn Research Institute, Bangkok 10210, Thailand (M.F.)
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok 10400, Thailand
- Program in Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok 10210, Thailand
| | - Ram Sasisekharan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charlermchai Artpradit
- Translational Research Unit, Chulabhorn Research Institute, Bangkok 10210, Thailand (M.F.)
| |
Collapse
|
2
|
Tan KW, Ji P, Zhou H, Zhang S, Zhou W. Further accelerating biologics development from DNA to IND: the journey from COVID-19 to non-COVID-19 programs. Antib Ther 2024; 7:96-104. [PMID: 38371952 PMCID: PMC10873266 DOI: 10.1093/abt/tbae001] [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: 10/17/2023] [Revised: 11/23/2023] [Accepted: 01/08/2024] [Indexed: 02/20/2024] Open
Abstract
The Coronavirus Disease (COVID-19) pandemic has spurred adoption of revolutionary initiatives by regulatory agencies and pharmaceutical industry worldwide to deliver therapeutic COVID-19 antibodies to patients at unprecedented speed. Among these, timeline of chemistry, manufacturing and control (CMC), which involves process development and manufacturing activities critical for the assurance of product quality and consistency before first-in-human clinical trials, was greatly reduced from typically 12-15 months (using clonal materials) to approximately 3 months (using non-clonal materials) in multiple cases. In this perspective, we briefly review the acceleration approaches published for therapeutic COVID-19 antibodies and subsequently discuss the applicability of these approaches to achieve investigational new drug (IND) timelines of ≤10 months in over 60 COVID-19 and non-COVID-19 programs performed at WuXi Biologics. We are of the view that, with demonstrated product quality and consistency, innovative approaches used for COVID-19 can be widely applied in all disease areas for greater speed to clinic.
Collapse
Affiliation(s)
- Kee Wee Tan
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Pengfei Ji
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Hang Zhou
- Bioprocess Research & Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Sam Zhang
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Weichang Zhou
- Biologics Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| |
Collapse
|
3
|
Balasubramanian S. Recombinant CHO Cell Pool Generation Using PiggyBac Transposon System. Methods Mol Biol 2024; 2810:137-146. [PMID: 38926277 DOI: 10.1007/978-1-0716-3878-1_9] [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: 06/28/2024]
Abstract
CHO cell pools with desirable characteristics of high titer and consistent product quality are useful for rapid production of recombinant proteins. Here, we describe the generation of CHO cell pools using the piggyBac transposon system for mediating gene integration. The method describes the co-transfection of cells with the donor plasmid (coding for the gene of interest) and the helper plasmid (coding for the transposase) using polyethyleneimine (PEI). This is followed by a genetic selection for the generation of a cell pool. The resulting cell pool can be used to start a batch or fed-batch culture. Alternatively, it can be used for generation of clonal cell lines or generation of cell banks for future use.
Collapse
Affiliation(s)
- Sowmya Balasubramanian
- Laboratory of Cellular Biotechnology (LBTC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Genentech, South San Francisco, CA, USA.
| |
Collapse
|
4
|
Barnard GC, Zhou M, Shen A, Yuk IH, Laird MW. Utilizing targeted integration CHO pools to potentially accelerate the GMP manufacturing of monoclonal and bispecific antibodies. Biotechnol Prog 2024; 40:e3399. [PMID: 37874920 DOI: 10.1002/btpr.3399] [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: 07/31/2023] [Revised: 10/03/2023] [Accepted: 10/08/2023] [Indexed: 10/26/2023]
Abstract
Monoclonal antibodies (mAbs) are effective therapeutic agents against many acute infectious diseases including COVID-19, Ebola, RSV, Clostridium difficile, and Anthrax. mAbs can therefore help combat a future pandemic. Unfortunately, mAb development typically takes years, limiting its potential to save lives during a pandemic. Therefore "pandemic mAb" timelines need to be shortened. One acceleration tool is "deferred cloning" and leverages new Chinese hamster ovary (CHO) technology based on targeted gene integration (TI). CHO pools, instead of CHO clones, can be used for Phase I/II clinical material production. A final CHO clone (producing the mAb with a similar product quality profile and preferably with a higher titer) can then be used for Phase III trials and commercial manufacturing. This substitution reduces timelines by ~3 months. We evaluated our novel CHO TI platform to enable deferred cloning. We created four unique CHO pools expressing three unique mAbs (mAb1, mAb2, and mAb3), and a bispecific mAb (BsAb1). We then performed single-cell cloning for mAb1 and mAb2, identifying three high-expressing clones from each pool. CHO pools and clones were inoculated side-by-side in ambr15 bioreactors. CHO pools yielded mAb titers as high as 10.4 g/L (mAb3) and 7.1 g/L (BsAb1). Subcloning yielded CHO clones expressing higher titers relative to the CHO pools while yielding similar product quality profiles. Finally, we showed that CHO TI pools were stable by performing a 3-month cell aging study. In summary, our CHO TI platform can increase the speed to clinic for a future "pandemic mAb."
Collapse
Affiliation(s)
- Gavin C Barnard
- Cell Culture and Bioprocess Operations, Genentech, South San Francisco, California, USA
| | - Michelle Zhou
- Cell Culture and Bioprocess Operations, Genentech, South San Francisco, California, USA
| | - Amy Shen
- Cell Culture and Bioprocess Operations, Genentech, South San Francisco, California, USA
| | - Inn H Yuk
- Cell Culture and Bioprocess Operations, Genentech, South San Francisco, California, USA
| | - Michael W Laird
- Cell Culture and Bioprocess Operations, Genentech, South San Francisco, California, USA
| |
Collapse
|
5
|
Delafosse L, Lord-Dufour S, Pelletier A, Perret S, Burlacu A, Ouimet M, Cass B, Joubert S, Stuible M, Durocher Y. Recombinant Protein Production from Stable CHO Cell Pools. Methods Mol Biol 2024; 2810:99-121. [PMID: 38926275 DOI: 10.1007/978-1-0716-3878-1_7] [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: 06/28/2024]
Abstract
The continuous improvement of expression platforms is necessary to respond to the increasing demand for recombinant proteins that are required to carry out structural or functional studies as well as for their characterization as biotherapeutics. While transient gene expression (TGE) in mammalian cells constitutes a rapid and well-established approach, non-clonal stably transfected cells, or "pools," represent another option, which is especially attractive when recurring productions of the same protein are required. From a culture volume of just a few liters, stable pools can provide hundreds of milligrams to gram quantities of high-quality secreted recombinant proteins.In this chapter, we describe a highly efficient and cost-effective procedure for the generation of Chinese Hamster Ovary cell stable pools expressing secreted recombinant proteins using commercially available serum-free media and polyethylenimine (PEI) as the transfection reagent. As a specific example of how this protocol can be applied, the production and downstream purification of recombinant His-tagged trimeric SARS-CoV-2 spike protein ectodomain (SmT1) are described.
Collapse
Affiliation(s)
- Laurence Delafosse
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Simon Lord-Dufour
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Alex Pelletier
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Sylvie Perret
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Alina Burlacu
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Manon Ouimet
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Brian Cass
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Simon Joubert
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada.
| |
Collapse
|
6
|
Maltais JS, Lord-Dufour S, Morasse A, Stuible M, Loignon M, Durocher Y. Repressing expression of difficult-to-express recombinant proteins during the selection process increases productivity of CHO stable pools. Biotechnol Bioeng 2023; 120:2840-2852. [PMID: 37232536 DOI: 10.1002/bit.28435] [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: 01/11/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023]
Abstract
More than half of licensed therapeutic recombinant proteins (r-proteins) are manufactured using constitutively-expressing, stably-transfected Chinese hamster ovary (CHO) clones. While constitutive CHO expression systems have proven their efficacy for the manufacturing of monoclonal antibodies, many next-generation therapeutics such as cytokines and bispecific antibodies as well as biological targets such as ectodomains of transmembrane receptors remain intrinsically challenging to produce. Herein, we exploited a cumate-inducible CHO platform allowing reduced expression of various classes of r-proteins during selection of stable pools. Following stable pool generation, fed-batch productions showed that pools generated without cumate (OFF-pools) were significantly more productive than pools selected in the presence of cumate (ON-pools) for 8 out of the 10 r-proteins tested, including cytokines, G-protein coupled receptors (GPCRs), the HVEM membrane receptor ectodomain, the multifunctional protein High Mobility Group protein B1 (HMGB1), as well as monoclonal and bispecific T-cell engager antibodies. We showed that OFF-pools contain a significantly larger proportion of cells producing high levels of r-proteins and that these cells tend to proliferate faster when expression is turned off, suggesting that r-protein overexpression imposes a metabolic burden on the cells. Cell viability was lower and pool recovery was delayed during selection of ON-pools (mimicking constitutive expression), suggesting that high producers were likely lost or overgrown by faster-growing, low-producing cells. We also observed a correlation between the expression levels of the GPCRs with Binding immunoglobulin Protein, an endoplasmic reticulum (ER) stress marker. Taken together, these data suggest that using an inducible system to minimize r-protein expression during stable CHO pool selection reduces cellular stresses, including ER stress and metabolic burden, leading to pools with greater frequency of high-expressing cells, resulting in improved volumetric productivity.
Collapse
Affiliation(s)
- Jean-Sébastien Maltais
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Simon Lord-Dufour
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Audrey Morasse
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Matthew Stuible
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Martin Loignon
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Yves Durocher
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
7
|
Joubert S, Stuible M, Lord-Dufour S, Lamoureux L, Vaillancourt F, Perret S, Ouimet M, Pelletier A, Bisson L, Mahimkar R, Pham PL, L Ecuyer-Coelho H, Roy M, Voyer R, Baardsnes J, Sauvageau J, St-Michael F, Robotham A, Kelly J, Acel A, Schrag JD, El Bakkouri M, Durocher Y. A CHO stable pool production platform for rapid clinical development of trimeric SARS-CoV-2 spike subunit vaccine antigens. Biotechnol Bioeng 2023. [PMID: 36987713 DOI: 10.1002/bit.28387] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/02/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023]
Abstract
Protein expression from stably transfected Chinese hamster ovary (CHO) clones is an established but time-consuming method for manufacturing therapeutic recombinant proteins. The use of faster, alternative approaches, such as non-clonal stable pools, has been restricted due to lower productivity and longstanding regulatory guidelines. Recently, the performance of stable pools has improved dramatically, making them a viable option for quickly producing drug substance for GLP-toxicology and early-phase clinical trials in scenarios such as pandemics that demand rapid production timelines. Compared to stable CHO clones which can take several months to generate and characterize, stable pool development can be completed in only a few weeks. Here, we compared the productivity and product quality of trimeric SARS-CoV-2 spike protein ectodomains produced from stable CHO pools or clones. Using a set of biophysical and biochemical assays we show that product quality is very similar and that CHO pools demonstrate sufficient productivity to generate vaccine candidates for early clinical trials. Based on these data, we propose that regulatory guidelines should be updated to permit production of early clinical trial material from CHO pools to enable more rapid and cost-effective clinical evaluation of potentially life-saving vaccines.
Collapse
Affiliation(s)
- Simon Joubert
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Simon Lord-Dufour
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Linda Lamoureux
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - François Vaillancourt
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Sylvie Perret
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Manon Ouimet
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Alex Pelletier
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Louis Bisson
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Rohan Mahimkar
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Phuong Lan Pham
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Helene L Ecuyer-Coelho
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Marjolaine Roy
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Robert Voyer
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Janelle Sauvageau
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Frank St-Michael
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Anna Robotham
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - John Kelly
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Andrea Acel
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Joseph D Schrag
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Majida El Bakkouri
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
8
|
Wang Y, Quan Q, Gleason C, Yu H, Peng L, Kang Y, Jiang L, Wu K, Pan J, Bao M, Zhu Q, Yi M, Fang M, Zheng Y, Qiu L, Xu B, Li X, Song J, Sun J, Zhang Z, Su Z, Lin J, Xie Y, Xu A, Song X, Huang C, Shen Z, Wang L, Song J. Accelerating the speed of innovative anti-tumor drugs to first-in-human trials incorporating key de-risk strategies. MAbs 2023; 15:2292305. [PMID: 38095560 DOI: 10.1080/19420862.2023.2292305] [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: 09/17/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
Pharmaceutical companies have recently focused on accelerating the timeline for initiating first-in-human (FIH) trials to allow quick assessment of biologic drugs. For example, a stable cell pool can be used to produce materials for the toxicology (Tox) study, reducing time to the clinic by 4-5 months. During the coronavirus disease 2019 (COVID-19) pandemic, the anti-COVID drugs timeline from DNA transfection to the clinical stage was decreased to 6 months using a stable pool to generate a clinical drug substrate (DS) with limited stability, virus clearance, and Tox study package. However, a lean chemistry, manufacturing, and controls (CMC) package raises safety and comparability risks and may leave extra work in the late-stage development and commercialization phase. In addition, whether these accelerated COVID-19 drug development strategies can be applied to non-COVID projects and established as a standard practice in biologics development is uncertain. Here, we present a case study of a novel anti-tumor drug in which application of "fast-to-FIH" approaches in combination with BeiGene's de-risk strategy achieved successful delivery of a complete CMC package within 10 months. A comprehensive comparability study demonstrated that the DS generated from a stable pool and a single-cell-derived master cell bank were highly comparable with regards to process performance, product quality, and potency. This accomplishment can be a blueprint for non-COVID drug programs that approach the pace of drug development during the pandemic, with no adverse impact on the safety, quality, and late-stage development of biologics.
Collapse
Affiliation(s)
- Yuqi Wang
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Quan Quan
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Camille Gleason
- Department of Regulatory Affairs CMC, BeiGene USA, Inc, San Mateo, CA, USA
| | - Helin Yu
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Lujia Peng
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Yanshen Kang
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Ling Jiang
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Kailun Wu
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Jie Pan
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Moxiyele Bao
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Qing Zhu
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Meiqi Yi
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Ming Fang
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Yue Zheng
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Ling Qiu
- Department of Technical Operation and Manufacturing, BeiGene (Guangzhou) Co. Ltd, Guangzhou, China
| | - Bin Xu
- Department of Technical Operation and Manufacturing, BeiGene (Guangzhou) Co. Ltd, Guangzhou, China
| | - Xiang Li
- Department of Technical Operation and Manufacturing, BeiGene (Guangzhou) Co. Ltd, Guangzhou, China
| | - Jinfeng Song
- Department of Technical Operation and Manufacturing, BeiGene (Guangzhou) Co. Ltd, Guangzhou, China
| | - Jiamu Sun
- Department of Regulatory Affairs CMC, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Zheng Zhang
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Zijun Su
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Jara Lin
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Yuanyuan Xie
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - April Xu
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Xiling Song
- Department of Regulatory Affairs CMC, BeiGene USA, Inc, San Mateo, CA, USA
| | - Chichi Huang
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Zhirong Shen
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Lai Wang
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| | - Jing Song
- Department of Research and Development, BeiGene (Beijing) Co. Ltd, Beijing, China
| |
Collapse
|
9
|
Tan KW, Ji P, Qian Z, Gao Q, Wang S, Li Q, Gu M, Zhang Q, Hou C, Huang Y, Lian D, Wang J, Zhang Z, Zhang S, Wu J, Zhou W. Rapidly accelerated development of neutralizing COVID-19 antibodies by reducing cell line and CMC development timelines. Biotechnol Bioeng 2022:10.1002/bit.28302. [PMID: 36482495 PMCID: PMC9877800 DOI: 10.1002/bit.28302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Since the Coronavirus Disease 2019 (COVID-19) outbreak, unconventional cell line development (CLD) strategies have been taken to enable development of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-neutralizing antibodies at expedited speed. We previously reported a novel chemistry, manufacturing, and control (CMC) workflow and demonstrated a much-shortened timeline of 3-6 months from DNA to investigational new drug (IND) application. Hereafter, we have incorporated this CMC strategy for many SARS-CoV-2-neutralizing antibody programs at WuXi Biologics. In this paper, we summarize the accelerated development of a total of seven antibody programs, some of which have received emergency use authorization approval in less than 2 years. Stable pools generated under good manufacturing practice (GMP) conditions consistently exhibited similar productivity and product quality at different scales and batches, enabling rapid initiation of phase I clinical trials. Clones with comparable product quality as parental pools were subsequently screened and selected for late-stage development and manufacturing. Moreover, a preliminary stability study plan was devised to greatly reduce the time required for final clone determination and next-generation sequencing-based viral testing was implemented to support rapid conditional release of the master cell bank for GMP production. The successful execution of these COVID-19 programs relies on our robust, fit for purpose, and continuously improving CLD platform. The speed achieved for pandemic-related biologics development may innovate typical biologics development timelines and become a new standard in the industry.
Collapse
Affiliation(s)
- Kee Wee Tan
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Pengfei Ji
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Zichen Qian
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Qiao Gao
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Shuai Wang
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Qin Li
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Mingzhu Gu
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Qi Zhang
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Chengjian Hou
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Yang Huang
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Dujuan Lian
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Junghao Wang
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Zheng Zhang
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Sam Zhang
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Jiansheng Wu
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| | - Weichang Zhou
- WuXi Biologics, Waigaoqiao Free Trade ZoneShanghaiChina
| |
Collapse
|
10
|
Kelley B, De Moor P, Douglas K, Renshaw T, Traviglia S. Monoclonal antibody therapies for COVID-19: lessons learned and implications for the development of future products. Curr Opin Biotechnol 2022; 78:102798. [PMID: 36179406 PMCID: PMC9436891 DOI: 10.1016/j.copbio.2022.102798] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 07/29/2022] [Accepted: 08/22/2022] [Indexed: 12/14/2022]
Abstract
Several companies were authorized to treat COVID-19 patients with monoclonal antibodies within 1-2 years of the start of the pandemic. These products were discovered, developed, manufactured, clinically tested, and approved under emergency-use authorization at unprecedented speed. Pandemic urgency led to novel development approaches that reduced the time to clinical trials by 75% or more without creating unacceptable patient or product-safety risks. Hundreds of thousands of patients now benefit from these therapeutics that have reduced the rates of hospitalization and death. The chemistry, manufacturing, and control development strategies set a new precedent of speed, safety, and demonstrated clinical benefit and will likely have a lasting impact on the development of future monoclonal antibody therapies for not only infectious diseases but also for oncology, inflammation, and rare diseases.
Collapse
Affiliation(s)
- Brian Kelley
- Vir Biotechnology, Inc., San Francisco, CA, USA.
| | | | | | | | | |
Collapse
|
11
|
Schmieder V, Fieder J, Drerup R, Gutierrez EA, Guelch C, Stolzenberger J, Stumbaum M, Mueller VS, Higel F, Bergbauer M, Bornhoefft K, Wittner M, Gronemeyer P, Braig C, Huber M, Reisenauer-Schaupp A, Mueller MM, Schuette M, Puengel S, Lindner B, Schmidt M, Schulz P, Fischer S. Towards maximum acceleration of monoclonal antibody development: Leveraging transposase-mediated cell line generation to enable GMP manufacturing within 3 months using a stable pool. J Biotechnol 2022; 349:53-64. [PMID: 35341894 DOI: 10.1016/j.jbiotec.2022.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/25/2022] [Accepted: 03/20/2022] [Indexed: 01/24/2023]
Abstract
In recent years, acceleration of development timelines has become a major focus within the biopharmaceutical industry to bring innovative therapies faster to patients. However, in order to address a high unmet medical need even faster further acceleration potential has to be identified to transform "speed-to-clinic" concepts into "warp-speed" development programs. Recombinant Chinese hamster ovary (CHO) cell lines are the predominant expression system for monoclonal antibodies (mAbs) and are routinely generated by random transgene integration (RTI) of the genetic information into the host cell genome. This process, however, exhibits considerable challenges such as the requirement for a time-consuming clone screening process to identify a suitable clonally derived manufacturing cell line. Hence, RTI represents an error prone and tedious method leading to long development timelines until availability of Good Manufacturing Practice (GMP)-grade drug substance (DS). Transposase-mediated semi-targeted transgene integration (STI) has been recently identified as a promising alternative to RTI as it allows for a more rapid generation of high-performing and stable production cell lines. In this report, we demonstrate how a STI technology was leveraged to develop a very robust DS manufacturing process based on a stable pool cell line at unprecedented pace. Application of the novel strategy resulted in the manufacturing of GMP-grade DS at 2,000 L scale in less than three months paving the way for a start of Phase I clinical trials only six months after transfection. Finally, using a clonally derived production cell line, which was established from the parental stable pool, we were able to successfully implement a process with an increased mAb titer of up to 5 g per liter at the envisioned commercial scale (12,000 L) within eight months.
Collapse
Affiliation(s)
- Valerie Schmieder
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Juergen Fieder
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Raphael Drerup
- Early Stage Bioprocess Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Erik Arango Gutierrez
- Early Stage Bioprocess Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Carina Guelch
- Late Stage Upstream Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Jessica Stolzenberger
- Late Stage Downstream Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Mihaela Stumbaum
- Early Stage Pharmaceutical Development, Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Volker Steffen Mueller
- Early Stage Analytics, Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Fabian Higel
- Early Stage Analytics, Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Martin Bergbauer
- Late Stage Analytics, Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Kim Bornhoefft
- Characterization Technologies, Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Manuel Wittner
- Global CMC Experts NBE, Global Quality Development, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Petra Gronemeyer
- Cell Banking & Inoculum, Focused Factory CS&T, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Christian Braig
- CST Transfer, Focused Factory CS&T, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Michaela Huber
- Process Transfer Cell Culture, Focused Factory Drug Substance, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Anita Reisenauer-Schaupp
- R&D PM NBE, Global R&D Project Management and Development Strategies, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Markus Michael Mueller
- CMC PM Process Industrialization Germany, Global Biopharma CMC Project Mgmt&TechRA, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Mark Schuette
- Global Technology Management, Global Innovation & Alliance Management, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Sebastian Puengel
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Benjamin Lindner
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Moritz Schmidt
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Patrick Schulz
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Simon Fischer
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany.
| |
Collapse
|
12
|
Agostinetto R, Rossi M, Dawson J, Lim A, Simoneau MH, Boucher C, Valldorf B, Ross‐Gillespie A, Jardine JG, Sok D, Burton DR, Hassell T, Broly H, Palinsky W, Dupraz P, Feinberg M, Dey AK. Rapid cGMP manufacturing of COVID-19 monoclonal antibody using stable CHO cell pools. Biotechnol Bioeng 2021; 119:663-666. [PMID: 34796474 PMCID: PMC8652680 DOI: 10.1002/bit.27995] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/12/2021] [Accepted: 11/13/2021] [Indexed: 01/08/2023]
Abstract
Therapeutic proteins, including monoclonal antibodies, are typically manufactured using clonally derived, stable host cell lines, since consistent and predictable cell culture performance is highly desirable. However, selecting and preparing banks of stable clones takes considerable time, which inevitably extends overall development timelines for new therapeutics by delaying the start of subsequent activities, such as the scale-up of manufacturing processes. In the context of the coronavirus disease 2019 (COVID-19) pandemic, with its intense pressure for accelerated development strategies, we used a novel transposon-based Leap-In Transposase® system to rapidly generate high-titer stable pools and then used them directly for large scale-manufacturing of an anti-severe acute respiratory syndrome coronavirus 2 monoclonal antibody under cGMP. We performed the safety testing of our non-clonal cell bank, then used it to produce material at a 200L-scale for preclinical safety studies and formulation development work, and thereafter at 2000L scale for supply of material for a Phase 1 clinical trial. Testing demonstrated the comparability of critical product qualities between the two scales and, more importantly, that our final clinical trial product met all pre-set product quality specifications. The above expediated approach provided clinical trial material within 4.5 months, in comparison to 12-14 months for production of clinical trial material via the conventional approach.
Collapse
Affiliation(s)
| | - Mara Rossi
- MerckSerono S.p.A, Guidonia di MontecelloItaly
| | | | | | | | - Cyril Boucher
- Ares Trading SA/Merck SA SwitzerlandAubonneSwitzerland
| | | | | | - Joseph G. Jardine
- Department of Immunology and MicrobiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
- IAVINew YorkNew YorkUSA
- IAVI, Neutralizing Antibody Center, The Scripps Research InstituteLa JollaCaliforniaUSA
| | - Devin Sok
- Department of Immunology and MicrobiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
- IAVINew YorkNew YorkUSA
- IAVI, Neutralizing Antibody Center, The Scripps Research InstituteLa JollaCaliforniaUSA
| | - Dennis R. Burton
- Department of Immunology and MicrobiologyThe Scripps Research InstituteLa JollaCaliforniaUSA
- IAVI, Neutralizing Antibody Center, The Scripps Research InstituteLa JollaCaliforniaUSA
- Ragon Institute of MGH, MIT and HarvardCambridgeMassachusettsUSA
| | | | - Hervé Broly
- Ares Trading SA/Merck SA SwitzerlandAubonneSwitzerland
| | - Wolf Palinsky
- Ares Trading SA/Merck SA SwitzerlandAubonneSwitzerland
| | | | | | - Antu K. Dey
- IAVINew YorkNew YorkUSA
- Present address:
Antu K. Dey, GreenLight Biosciences Inc., 200 Boston Avenue, Suite 1000MedfordMassachusettsUSA
| |
Collapse
|
13
|
Kumar R, Guttman A, Rathore AS. Applications of capillary electrophoresis for biopharmaceutical product characterization. Electrophoresis 2021; 43:143-166. [PMID: 34591322 DOI: 10.1002/elps.202100182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/07/2021] [Accepted: 09/23/2021] [Indexed: 12/16/2022]
Abstract
Capillary electrophoresis (CE), after being introduced several decades ago, has carved out a niche for itself in the field of analytical characterization of biopharmaceutical products. It does not only offer fast separation, high resolution in miniaturized format, but equally importantly represents an orthogonal separation mechanism to high-performance liquid chromatography. Therefore, it is not surprising that CE-based methods can be found in all major pharmacopoeias and are recommended for the analysis of biopharmaceutical products during process development, characterization, quality control, and release testing. Different separation formats of CE, such as capillary gel electrophoresis, capillary isoelectric focusing, and capillary zone electrophoresis are widely used for size and charge heterogeneity characterization as well as purity and stability testing of therapeutic proteins. Hyphenation of CE with MS is emerging as a promising bioanalytical tool to assess the primary structure of therapeutic proteins along with any impurities. In this review, we confer the latest developments in capillary electrophoresis, used for the characterization of critical quality attributes of biopharmaceutical products covering the past 6 years (2015-2021). Monoclonal antibodies, due to their significant share in the market, have been given prioritized coverage.
Collapse
Affiliation(s)
- Ramesh Kumar
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, India
| | - Andras Guttman
- Horváth Csaba Memorial Laboratories of Bioseparation Sciences, Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Translational Glycomics Group, Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Veszprem, Hungary
| | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, India
| |
Collapse
|
14
|
McGovern ĀT, Salisbury CM, Nyberg GB. The pandemic and resilience for the future: AccBio 2021. Biotechnol Prog 2021; 38:e3207. [PMID: 34463436 PMCID: PMC8646774 DOI: 10.1002/btpr.3207] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/06/2022]
Abstract
The year 2020 brought the onslaught of a global crisis in the form of the COVID-19 pandemic. While nearly every facet of everyday life and work was impacted by the pandemic, the biopharmaceutical industry found silver linings in innovation, partnership, and resiliency, all of which contributed to unprecedented speed in developing and delivering vaccines and therapies. The 7th International Conference on Accelerating Biopharmaceutical Development (AccBio 2021) brought together industry leaders to share experiences from the past year and discuss how lessons learned from the pandemic can be carried forward into the future of biopharmaceutical development. Presenters highlighted examples such as introducing biotherapeutics derived from non-clonal cell pools into the clinic, developing modular or platform technologies, and taking novel risks, among others. These strategies for enabling speed to clinic and launch, as well as for sustaining a robust supply chain, are likely to be integrated into future programs to ensure biomanufacturing resiliency and get medicines to patients faster than pre-pandemic times.
Collapse
Affiliation(s)
- Āine T McGovern
- Pharma Technical Development US Biologics, Genentech, South San Francisco, California, USA
| | - Cleo M Salisbury
- Pharma Technical Development US Biologics, Genentech, South San Francisco, California, USA
| | - Gregg B Nyberg
- Biologics Process Research & Development, Merck & Co., Inc., Kenilworth, New Jersey, USA
| |
Collapse
|
15
|
Diep J, Le H, Le K, Zasadzinska E, Tat J, Yam P, Zastrow R, Gomez N, Stevens J. Microfluidic chip-based single-cell cloning to accelerate biologic production timelines. Biotechnol Prog 2021; 37:e3192. [PMID: 34323013 PMCID: PMC9285370 DOI: 10.1002/btpr.3192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/06/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022]
Abstract
Cell line development (CLD) represents a critical, yet time‐consuming, step in the biomanufacturing process as significant resources are devoted to the scale‐up and screening of several hundreds to thousands of single‐cell clones. Typically, transfected pools are fully recovered from selection and characterized for growth, productivity, and product quality to identify the best pools suitable for single‐cell cloning (SCC) using limiting dilution or fluorescence‐activated cell sorting (FACS). Here we report the application of the Berkeley Lights Beacon Instrument (BLI) in an early SCC process to accelerate the CLD timeline. Transfected pools were single‐cell cloned when viabilities reached greater than 85% or during selection when viabilities were less than 30%. Clones isolated from these accelerated processes exhibited comparable growth, productivity, and product quality to those derived from a standard CLD process and fit into an existing manufacturing platform. With these approaches, up to a 30% reduction in the overall CLD timeline was achieved. Furthermore, early process‐derived clones demonstrated equivalent long‐term stability compared with standard process‐derived clones over 50 population doubling levels (PDLs). Taken together, the data supported early SCC on the BLI as an attractive approach to reducing the standard CLD timeline while still identifying clones with acceptable manufacturability.
Collapse
Affiliation(s)
- Jonathan Diep
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Huong Le
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Kim Le
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Ewelina Zasadzinska
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Jasmine Tat
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Pheng Yam
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Ryan Zastrow
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Natalia Gomez
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| | - Jennitte Stevens
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California, USA
| |
Collapse
|
16
|
Zhang Z, Chen J, Wang J, Gao Q, Ma Z, Xu S, Zhang L, Cai J, Zhou W. Reshaping cell line development and CMC strategy for fast responses to pandemic outbreak. Biotechnol Prog 2021; 37:e3186. [PMID: 34148295 DOI: 10.1002/btpr.3186] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/12/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022]
Abstract
The global pandemic outbreak COVID-19 (SARS-COV-2), has prompted many pharmaceutical companies to develop vaccines and therapeutic biologics for its prevention and treatment. Most of the therapeutic biologics are common human IgG antibodies, which were identified by next-generation sequencing (NGS) with the B cells from the convalescent patients. To fight against pandemic outbreaks like COVID-19, biologics development strategies need to be optimized to speed up the timeline. Since the advent of therapeutic biologics, strategies of transfection and cell line selection have been continuously improved for greater productivity and efficiency. NGS has also been implemented for accelerated cell bank testing. These recent advances enable us to rethink and reshape the chemistry, manufacturing, and controls (CMC) strategy in order to start supplying Good Manufacturing Practices (GMP) materials for clinical trials as soon as possible. We elucidated an accelerated CMC workflow for biologics, including using GMP-compliant pool materials for phase I clinical trials, selecting the final clone with product quality similar to that of phase I materials for late-stage development and commercial production.
Collapse
Affiliation(s)
- Zheng Zhang
- Waigaoqiao Free Trade Zone, WuXi Biologics, Shanghai, China
| | - Ji Chen
- Waigaoqiao Free Trade Zone, WuXi Biologics, Shanghai, China
| | - Junghao Wang
- Waigaoqiao Free Trade Zone, WuXi Biologics, Shanghai, China
| | - Qiao Gao
- Waigaoqiao Free Trade Zone, WuXi Biologics, Shanghai, China
| | - Zhujun Ma
- Waigaoqiao Free Trade Zone, WuXi Biologics, Shanghai, China
| | - Shurong Xu
- Waigaoqiao Free Trade Zone, WuXi Biologics, Shanghai, China
| | - Li Zhang
- Waigaoqiao Free Trade Zone, WuXi Biologics, Shanghai, China
| | - Jill Cai
- Waigaoqiao Free Trade Zone, WuXi Biologics, Shanghai, China
| | - Weichang Zhou
- Waigaoqiao Free Trade Zone, WuXi Biologics, Shanghai, China
| |
Collapse
|
17
|
Rajendran S, Balasubramanian S, Webster L, Lee M, Vavilala D, Kulikov N, Choi J, Tang C, Hunter M, Wang R, Kaur H, Karunakaran S, Sitaraman V, Minshull J, Boldog F. Accelerating and de-risking CMC development with transposon-derived manufacturing cell lines. Biotechnol Bioeng 2021; 118:2301-2311. [PMID: 33704772 PMCID: PMC8252637 DOI: 10.1002/bit.27742] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 01/08/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022]
Abstract
The development of highly productive, genetically stable manufacturing cell lines is on the critical path to IND filing for protein-based biologic drugs. Here, we describe the Leap-In Transposase® platform, a novel transposon-based mammalian (e.g., Chinese hamster ovary) cell line development system that produces high-titer stable pools with productivity and product quality attributes that are highly comparable to clones that are subsequently derived therefrom. The productivity distributions of clones are strongly biased toward high producers, and genetic and expression stability is consistently high. By avoiding the poor integration rates, concatemer formation, detrimental transgene recombination, low average expression level, unpredictable product quality, and inconsistent genetic stability characteristic of nonhomologous recombination methods, Leap-In provides several opportunities to de-risk programs early and reduce timelines and resources.
Collapse
Affiliation(s)
- Sowmya Rajendran
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Sowmya Balasubramanian
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Lynn Webster
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Maggie Lee
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Divya Vavilala
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Nicolay Kulikov
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Jessica Choi
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Calvin Tang
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Molly Hunter
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Rebecca Wang
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Harpreet Kaur
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Surya Karunakaran
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Varsha Sitaraman
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Jeremy Minshull
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| | - Ferenc Boldog
- Cell Line DevelopmentProtein Purification and Protein Analytical Departments of ATUM, Inc.NewarkCaliforniaUSA
| |
Collapse
|
18
|
Dong E, Lam C, Tang D, Louie S, Yim M, Williams AJ, Sawyer W, Yip S, Carver J, AlBarakat A, Tsukuda J, Snedecor B, Misaghi S. Concurrent transfection of randomized transgene configurations into targeted integration CHO host is an advantageous and cost-effective method for expression of complex molecules. Biotechnol J 2020; 16:e2000230. [PMID: 33259700 DOI: 10.1002/biot.202000230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022]
Abstract
Complex recombinant proteins are increasingly desired as potential therapeutic options for many disease indications and are commonly expressed in the mammalian Chinese hamster ovary (CHO) cells. Generally, stoichiometric expression and proper folding of all subunits of a complex recombinant protein are required to achieve the desired titers and product qualities for a complex molecule. Targeted integration (TI) cell line development (CLD), which entails the insertion of the desired transgene(s) into a predefined landing-pad in the CHO genome, enables the generation of a homogeneous pool of cells from which clonally stable and high titer clones can be isolated with minimal screening efforts. Despite these advantages, using a single transgene(s) configuration with predetermined gene dosage might not be adequate for the expression of complex molecules. The goal of this study is to develop a method for seamless screening of many vector configurations in a single TI CLD attempt. As testing vector configurations in transient expression systems is not predictive of protein expression in the stable cell lines and parallel TI CLDs with different transgene configurations is resource-intensive, we tested the concept of randomized configuration targeted integration (RCTI) CLD approach for expression of complex molecules. RCTI allows simultaneous transfection of multiple vector configurations, encoding a complex molecule, to generate diverse TI clones each with a single transgene configuration but clone specific productivity and product qualities. Our findings further revealed a direct correlation between transgenes' configuration/copy-number and titer/product quality of the expressed proteins. RCTI CLD enabled, with significantly fewer resources, seamless isolation of clones with comparable titers and product quality attributes to that of several parallel standard TI CLDs. Therefore, RCTI introduces randomness to the TI CLD platform while maintaining all the advantages, such as clone stability and reduced sequence variant levels, that the TI system has to offer.
Collapse
Affiliation(s)
- Emily Dong
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Cynthia Lam
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Danming Tang
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Salina Louie
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Mandy Yim
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Ambrose J Williams
- Purification Development Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - William Sawyer
- Biochemical and Cellular Pharmacology Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Shirley Yip
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Joseph Carver
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Ali AlBarakat
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Joni Tsukuda
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Brad Snedecor
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| | - Shahram Misaghi
- Cell Culture and Bioprocess Operations Department, Genentech, Inc. 1 DNA Way, South San Francisco, California, USA
| |
Collapse
|
19
|
Abstract
The time from discovery to proof-of-concept trials could be reduced to 5–6 months from a traditional timeline of 10–12 months.
Collapse
|
20
|
Bolisetty P, Tremml G, Xu S, Khetan A. Enabling speed to clinic for monoclonal antibody programs using a pool of clones for IND-enabling toxicity studies. MAbs 2020; 12:1763727. [PMID: 32449878 PMCID: PMC7531531 DOI: 10.1080/19420862.2020.1763727] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/15/2020] [Accepted: 04/29/2020] [Indexed: 12/23/2022] Open
Abstract
The importance of speed to clinic for medicines that may address unmet medical needs puts pressure on product development timelines. Historically, both toxicology and first-in-human clinical materials are generated using the same clonal-derived cells to ensure safety and minimize any development risks. However, cell line development with single cell cloning is time consuming, and aggravated by the time needed to screen for a lead clone based on cell line stability and manufacturability. In order to achieve faster timelines, we have used pools of up to six clones for earlier production of drug substance for regulatory filing-enabling toxicology studies, and then the final single clone was selected for production of clinical materials. This approach was enabled by using platform processes across all stages of early development, including expression vectors, host cell lines, media, and production processes. Through comprehensive cell culture and product quality analysis, we demonstrated that the toxicology material was representative of the clinical material for all six monoclonal antibody programs evaluated. Our extensive development experience further confirmed that using a pool of clones for toxicology material generation is a reliable approach to shorten the early development timeline.
Collapse
Affiliation(s)
| | - Gabi Tremml
- Biologics Development, Bristol Myers Squibb Co, New Brunswick, NJ, USA
| | - Sen Xu
- Biologics Development, Bristol Myers Squibb Co, New Brunswick, NJ, USA
| | - Anurag Khetan
- Biologics Development, Bristol Myers Squibb Co, New Brunswick, NJ, USA
| |
Collapse
|
21
|
Welch JT, Arden NS. Considering “clonality”: A regulatory perspective on the importance of the clonal derivation of mammalian cell banks in biopharmaceutical development. Biologicals 2019; 62:16-21. [DOI: 10.1016/j.biologicals.2019.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 12/23/2022] Open
|
22
|
Stuible M, van Lier F, Croughan MS, Durocher Y. Beyond preclinical research: production of CHO-derived biotherapeutics for toxicology and early-phase trials by transient gene expression or stable pools. Curr Opin Chem Eng 2018. [DOI: 10.1016/j.coche.2018.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Balasubramanian S, Peery RB, Minshull J, Lee M, White R, Kelly RM, Barnard GC. Generation of High Expressing Chinese Hamster Ovary Cell Pools Using the Leap-In Transposon System. Biotechnol J 2018; 13:e1700748. [DOI: 10.1002/biot.201700748] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 03/29/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Sowmya Balasubramanian
- Bioprocess Research and Development; Eli Lilly and Company; LTC-North, 1200 Kentucky AvenueIndianapolis IN 46221 USA
| | - Robert B. Peery
- Biotechnology Discovery Research; Lilly Research Laboratories; Eli Lilly and Company; Lilly Corporate Center; Indianapolis IN 46225 USA
| | | | - Maggie Lee
- ATUM; 37950 Central CtNewark CA 94560 USA
| | - Regina White
- Biotechnology Discovery Research; Lilly Research Laboratories; Eli Lilly and Company; Lilly Corporate Center; Indianapolis IN 46225 USA
| | - Ronan M. Kelly
- Bioprocess Research and Development; Eli Lilly and Company; LTC-North, 1200 Kentucky AvenueIndianapolis IN 46221 USA
| | - Gavin C. Barnard
- Biotechnology Discovery Research; Lilly Research Laboratories; Eli Lilly and Company; Lilly Corporate Center; Indianapolis IN 46225 USA
| |
Collapse
|
24
|
Abstract
CHO cell pools with desirable characteristics of high titer and consistent product quality are useful for rapid production of recombinant proteins. Here we describe the generation of CHO cell pools using the piggyBac transposon system for mediating gene integration. The method describes the co-transfection of cells with the donor plasmid (coding for the gene of interest) and the helper plasmid (coding for the transposase) using polyethyleneimine (PEI). This is followed by a genetic selection for the generation of a cell pool. The resulting cell pool can be used to start a batch or fed-batch culture. Alternatively it can be used for generation of clonal cell lines or generation of cell banks for future use.
Collapse
Affiliation(s)
- Sowmya Balasubramanian
- Laboratory of Cellular Biotechnology (LBTC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland. .,ATUM, Newark, CA, USA.
| |
Collapse
|