1
|
Zeh N, Schmidt M, Schulz P, Fischer S. The new frontier in CHO cell line development: From random to targeted transgene integration technologies. Biotechnol Adv 2024; 75:108402. [PMID: 38950872 DOI: 10.1016/j.biotechadv.2024.108402] [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: 02/24/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/03/2024]
Abstract
Cell line development represents a crucial step in the development process of a therapeutic glycoprotein. Chinese hamster ovary (CHO) cells are the most frequently employed mammalian host cell system for the industrial manufacturing of biologics. The predominant application of CHO cells for heterologous recombinant protein expression lies in the relative simplicity of stably introducing ectopic DNA into the CHO host cell genome. Since CHO cells were first used as expression host for the industrial production of biologics in the late 1980s, stable genomic transgene integration has been achieved almost exclusively by random integration. Since then, random transgene integration had become the gold standard for generating stable CHO production cell lines due to a lack of viable alternatives. However, it was eventually demonstrated that this approach poses significant challenges on the cell line development process such as an increased risk of inducing cell line instability. In recent years, significant discoveries of new and highly potent (semi)-targeted transgene integration systems have paved the way for a technological revolution in the cell line development sector. These advanced methodologies comprise the application of transposase-, recombinase- or Cas9 nuclease-mediated site-specific genomic integration techniques, which enable a scarless transfer of the transgene expression cassette into transcriptionally active loci within the host cell genome. This review summarizes recent advancements in the field of transgene integration technologies for CHO cell line development and compare them to the established random integration approach. Moreover, advantages and limitations of (semi)-targeted integration techniques are discussed, and benefits and opportunities for the biopharmaceutical industry are outlined.
Collapse
Affiliation(s)
- Nikolas Zeh
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH and Co.KG, Biberach an der Riss, Germany
| | - Moritz Schmidt
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH and Co.KG, Biberach an der Riss, Germany
| | - Patrick Schulz
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH and Co.KG, Biberach an der Riss, Germany
| | - Simon Fischer
- Cell Line Development, Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH and Co.KG, Biberach an der Riss, Germany.
| |
Collapse
|
2
|
Tu B, Lin Z, Moore J, Krishnan Sekaran A, Wesley MJ, Mao DY, Gibson M, Lai WC, Boggs J, Slowik T, Perez-Gelvez YNC, Bonn R, Rae T, Minshull J, Boldog F, Sitaraman V, Muerhoff S, Hemken P. Recombinant Antibody-Producing Stable CHOK1 Pool Stability Study. Monoclon Antib Immunodiagn Immunother 2024; 43:119-126. [PMID: 39034896 DOI: 10.1089/mab.2024.0008] [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: 07/23/2024] Open
Abstract
Mammalian cell line stability is an important consideration when establishing a biologics manufacturing process in the biopharmaceutical and in vitro diagnostics (IVD) industries. Traditional Chinese hamster ovary (CHO) cell line development methods use a random integration approach that requires transfection, selection, optional amplification, screenings, and single-cell cloning to select clones with acceptable productivity, product quality, and genetic stability. Site-specific integration reduces these disadvantages, and new technologies have been developed to mitigate risks associated with genetic instability. In this study, we applied the Leap-In® transposase-mediated expression system from ATUM to generate stable CHOK1 pools for the production of four recombinant antibody reagents for IVD immunoassays. CHO cell line stability is defined by consistent antibody production over time. Three of the CHOK1 pools maintained productivity suitable for manufacturing, with high antibody yields. The productivity of the remaining CHOK1 pool decreased over time; however, derivative clones showed acceptable stability. l-glutamine had variable effects on CHOK1 cell line or stable pool stability and significantly affected antibody product titer. Compared with traditional random integration methods, the ATUM Leap-In system can reduce the time needed to develop new immunoassays by using semi site-specific integration to generate high-yield stable pools that meet manufacturing stability requirements.
Collapse
Affiliation(s)
- Bailin Tu
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Zhihong Lin
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Jeff Moore
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Archana Krishnan Sekaran
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Miranda J Wesley
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - De Yu Mao
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Mark Gibson
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Wan-Ching Lai
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - John Boggs
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Thomas Slowik
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Yeni Natalia C Perez-Gelvez
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Ryan Bonn
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Tracey Rae
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | | | | | | | - Scott Muerhoff
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Philip Hemken
- Biologics Discovery and Design, Abbott Diagnostics Division, Abbott Laboratories, Abbott Park, Illinois, USA
| |
Collapse
|
3
|
Lenser M, Ngo HG, Sarrafha L, Rajendra Y. Evaluation of two transposases for improving expression of recombinant proteins in Chinese hamster ovary cell stable pools by co-transfection and supertransfection approaches. Biotechnol Prog 2024:e3496. [PMID: 39016635 DOI: 10.1002/btpr.3496] [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: 03/26/2024] [Revised: 06/13/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024]
Abstract
Transposons are genetic elements capable of cutting and pasting genes of interest via the action of a transposase and offer many advantages over random or targeted integration of DNA in the creation of Chinese hamster ovary (CHO) cell lines for recombinant protein expression. Unique transposases have different recognition sites, allowing multiple transposases to be co-transfected together. They also allow for supertransfection (transfection on a previously transfected pool or cell line) with a second transposase to integrate additional copies of the same gene or an additional gene without disruption of the previously integrated DNA which to our knowledge has not been previously described in literature. Two fluorescent proteins, EGFP and tagRFP657, were either co-transfected or supertransfected into CHO cells using two unique transposases and showed high expression efficiency with similar expression levels (measured as mean fluorescence intensity), regardless of whether the genes were co-transfected or supertransfected onto an existing stable pool. Additionally, dual selection of the genes, both in the absence of L-glutamine and the presence of puromycin, led to higher expression levels than single selection alone. These results demonstrate that supertransfection using unique transposases could be a useful strategy for increasing titers of existing cell lines or for overexpressing helper (non-therapeutic) genes to improve expression and/or product quality of existing pools and cell lines, potentially saving significant time and resources.
Collapse
Affiliation(s)
- Melina Lenser
- Bioprocess Development, Technical Operations, Denali Therapeutics, Inc., South San Francisco, California, USA
| | - Hanh Giai Ngo
- Bioprocess Development, Technical Operations, Denali Therapeutics, Inc., South San Francisco, California, USA
| | - Lily Sarrafha
- Discovery Biology, Denali Therapeutics, Inc., South San Francisco, California, USA
| | - Yashas Rajendra
- Bioprocess Development, Technical Operations, Denali Therapeutics, Inc., South San Francisco, California, USA
| |
Collapse
|
4
|
Phan T, Ye Q, Stach C, Lin YC, Cao H, Bowen A, Langlois RA, Hu WS. Synthetic Cell Lines for Inducible Packaging of Influenza A Virus. ACS Synth Biol 2024; 13:546-557. [PMID: 38259154 PMCID: PMC10878389 DOI: 10.1021/acssynbio.3c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024]
Abstract
Influenza A virus (IAV) is a negative-sense RNA virus that causes seasonal infections and periodic pandemics, inflicting huge economic and human costs on society. The current production of influenza virus for vaccines is initiated by generating a seed virus through the transfection of multiple plasmids in HEK293 cells followed by the infection of seed viruses into embryonated chicken eggs or cultured mammalian cells. We took a system design and synthetic biology approach to engineer cell lines that can be induced to produce all viral components except hemagglutinin (HA) and neuraminidase (NA), which are the antigens that specify the variants of IAV. Upon the transfection of HA and NA, the cell line can produce infectious IAV particles. RNA-Seq transcriptome analysis revealed inefficient synthesis of viral RNA and upregulated expression of genes involved in host response to viral infection as potential limiting factors and offered possible targets for enhancing the productivity of the synthetic cell line. Overall, we showed for the first time that it was possible to create packaging cell lines for the production of a cytopathic negative-sense RNA virus. The approach allows for the exploitation of altered kinetics of the synthesis of viral components and offers a new method for manufacturing viral vaccines.
Collapse
Affiliation(s)
- Thu Phan
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Qian Ye
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
- State
Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Christopher Stach
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Yu-Chieh Lin
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Haoyu Cao
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Annika Bowen
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ryan A. Langlois
- Department
of Microbiology and Immunology, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Wei-Shou Hu
- Department
of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
5
|
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
|
6
|
Weidenbacher PAB, Sanyal M, Friedland N, Tang S, Arunachalam PS, Hu M, Kumru OS, Morris MK, Fontenot J, Shirreff L, Do J, Cheng YC, Vasudevan G, Feinberg MB, Villinger FJ, Hanson C, Joshi SB, Volkin DB, Pulendran B, Kim PS. A ferritin-based COVID-19 nanoparticle vaccine that elicits robust, durable, broad-spectrum neutralizing antisera in non-human primates. Nat Commun 2023; 14:2149. [PMID: 37069151 PMCID: PMC10110616 DOI: 10.1038/s41467-023-37417-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/16/2023] [Indexed: 04/19/2023] Open
Abstract
While the rapid development of COVID-19 vaccines has been a scientific triumph, the need remains for a globally available vaccine that provides longer-lasting immunity against present and future SARS-CoV-2 variants of concern (VOCs). Here, we describe DCFHP, a ferritin-based, protein-nanoparticle vaccine candidate that, when formulated with aluminum hydroxide as the sole adjuvant (DCFHP-alum), elicits potent and durable neutralizing antisera in non-human primates against known VOCs, including Omicron BQ.1, as well as against SARS-CoV-1. Following a booster ~one year after the initial immunization, DCFHP-alum elicits a robust anamnestic response. To enable global accessibility, we generated a cell line that can enable production of thousands of vaccine doses per liter of cell culture and show that DCFHP-alum maintains potency for at least 14 days at temperatures exceeding standard room temperature. DCFHP-alum has potential as a once-yearly (or less frequent) booster vaccine, and as a primary vaccine for pediatric use including in infants.
Collapse
Affiliation(s)
- Payton A-B Weidenbacher
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Mrinmoy Sanyal
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Natalia Friedland
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Shaogeng Tang
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Mengyun Hu
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Ozan S Kumru
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | | | - Jane Fontenot
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Lisa Shirreff
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Jonathan Do
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Ya-Chen Cheng
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | | | | | - Francois J Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Carl Hanson
- California Department of Public Health, Richmond, CA, USA
| | - Sangeeta B Joshi
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - David B Volkin
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter S Kim
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| |
Collapse
|
7
|
Fernández-Quintero ML, Ljungars A, Waibl F, Greiff V, Andersen JT, Gjølberg TT, Jenkins TP, Voldborg BG, Grav LM, Kumar S, Georges G, Kettenberger H, Liedl KR, Tessier PM, McCafferty J, Laustsen AH. Assessing developability early in the discovery process for novel biologics. MAbs 2023; 15:2171248. [PMID: 36823021 PMCID: PMC9980699 DOI: 10.1080/19420862.2023.2171248] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/18/2023] [Indexed: 02/25/2023] Open
Abstract
Beyond potency, a good developability profile is a key attribute of a biological drug. Selecting and screening for such attributes early in the drug development process can save resources and avoid costly late-stage failures. Here, we review some of the most important developability properties that can be assessed early on for biologics. These include the influence of the source of the biologic, its biophysical and pharmacokinetic properties, and how well it can be expressed recombinantly. We furthermore present in silico, in vitro, and in vivo methods and techniques that can be exploited at different stages of the discovery process to identify molecules with liabilities and thereby facilitate the selection of the most optimal drug leads. Finally, we reflect on the most relevant developability parameters for injectable versus orally delivered biologics and provide an outlook toward what general trends are expected to rise in the development of biologics.
Collapse
Affiliation(s)
- Monica L. Fernández-Quintero
- Center for Molecular Biosciences Innsbruck (CMBI), Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Franz Waibl
- Center for Molecular Biosciences Innsbruck (CMBI), Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Victor Greiff
- Department of Immunology, University of Oslo, Oslo, Norway
| | - Jan Terje Andersen
- Department of Immunology, University of Oslo, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo, Oslo, Norway
| | | | - Timothy P. Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Bjørn Gunnar Voldborg
- National Biologics Facility, Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lise Marie Grav
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Sandeep Kumar
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Guy Georges
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Hubert Kettenberger
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Klaus R. Liedl
- Center for Molecular Biosciences Innsbruck (CMBI), Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innsbruck, Austria
| | - Peter M. Tessier
- Department of Chemical Engineering, Pharmaceutical Sciences and Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - John McCafferty
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Maxion Therapeutics, Babraham Research Campus, Cambridge, UK
| | - Andreas H. Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
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
|
Hall CA, Kravitz RH, Johnson KF, Sanek NA, Maiti P, Ziemba KR, Liu J, Andreev DO, Chrostowski VL, Collins IJ, Bleck GT. Shortening the biologics clinical timeline with a novel method for generating stable, high-producing cell pools and clones. Biotechnol Bioeng 2022. [PMID: 36582005 DOI: 10.1002/bit.28323] [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: 12/14/2022] [Accepted: 12/18/2022] [Indexed: 12/31/2022]
Abstract
Reducing drug development timelines is an industry-wide goal to bring medicines to patients in need more quickly. This was exemplified in the coronavirus disease 2019 pandemic where reducing development timelines had a direct impact on the number of lives lost to the disease. The use of drug substances produced using cell pools, as opposed to clones, has the potential to shorten development timelines. Toward this goal, we have developed a novel technology, GPEx® Lightning, that allows for rapid, reproducible, targeted recombination of transgenes into more than 200 Dock sites in the Chinese hamster ovary cell line genome. This allows for rapid production of high-expressing stable cell pools and clones that reach titers of 4-12 g/l in generic fed-batch production. These pools and clones are highly stable in both titer and glycosylation, showing strong similarities in glycosylation profiles.
Collapse
Affiliation(s)
- Chad A Hall
- Catalent Pharma Solutions, Somerset, New Jersey, USA
| | | | | | | | - Payel Maiti
- Catalent Pharma Solutions, Somerset, New Jersey, USA
| | | | - Jia Liu
- Catalent Pharma Solutions, Somerset, New Jersey, USA
| | | | | | - Ian J Collins
- Catalent Pharma Solutions, Somerset, New Jersey, USA
| | | |
Collapse
|
10
|
Weidenbacher PAB, Sanyal M, Friedland N, Tang S, Arunachalam PS, Hu M, Kumru OS, Morris MK, Fontenot J, Shirreff L, Do J, Cheng YC, Vasudevan G, Feinberg MB, Villinger FJ, Hanson C, Joshi SB, Volkin DB, Pulendran B, Kim PS. A ferritin-based COVID-19 nanoparticle vaccine that elicits robust, durable, broad-spectrum neutralizing antisera in non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.12.25.521784. [PMID: 36597527 PMCID: PMC9810210 DOI: 10.1101/2022.12.25.521784] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
While the rapid development of COVID-19 vaccines has been a scientific triumph, the need remains for a globally available vaccine that provides longer-lasting immunity against present and future SARS-CoV-2 variants of concern (VOCs). Here, we describe DCFHP, a ferritin-based, protein-nanoparticle vaccine candidate that, when formulated with aluminum hydroxide as the sole adjuvant (DCFHP-alum), elicits potent and durable neutralizing antisera in non-human primates against known VOCs, including Omicron BQ.1, as well as against SARS-CoV-1. Following a booster ∼one year after the initial immunization, DCFHP-alum elicits a robust anamnestic response. To enable global accessibility, we generated a cell line that can enable production of thousands of vaccine doses per liter of cell culture and show that DCFHP-alum maintains potency for at least 14 days at temperatures exceeding standard room temperature. DCFHP-alum has potential as a once-yearly booster vaccine, and as a primary vaccine for pediatric use including in infants.
Collapse
Affiliation(s)
- Payton A.-B. Weidenbacher
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Mrinmoy Sanyal
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Natalia Friedland
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Shaogeng Tang
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Prabhu S. Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Mengyun Hu
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Ozan S. Kumru
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | | | - Jane Fontenot
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Lisa Shirreff
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Jonathan Do
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Ya-Chen Cheng
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | | | | | - Francois J. Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Carl Hanson
- California Department of Public Health, Richmond, CA, USA
| | - Sangeeta B. Joshi
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - David B. Volkin
- Vaccine Analytics and Formulation Center, Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter S. Kim
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, United States
| |
Collapse
|
11
|
Wang Y, Qiu H, Minshull J, Tam W, Hu X, Mieczkowski C, Zheng W, Chu C, Liu W, Boldog F, Gustafsson C, Gries JM, Xu W. An innovative platform to improve asymmetric bispecific antibody assembly, purity, and expression level in stable pool and cell line development. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
12
|
Wei M, Mi CL, Jing CQ, Wang TY. Progress of Transposon Vector System for Production of Recombinant Therapeutic Proteins in Mammalian Cells. Front Bioeng Biotechnol 2022; 10:879222. [PMID: 35600890 PMCID: PMC9114503 DOI: 10.3389/fbioe.2022.879222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, mammalian cells have become the primary host cells for the production of recombinant therapeutic proteins (RTPs). Despite that the expression of RTPs in mammalian cells can be improved by directly optimizing or engineering the expression vectors, it is still influenced by the low stability and efficiency of gene integration. Transposons are mobile genetic elements that can be inserted and cleaved within the genome and can change their inserting position. The transposon vector system can be applied to establish a stable pool of cells with high efficiency in RTPs production through facilitating the integration of gene of interest into transcriptionally active sites under screening pressure. Here, the structure and optimization of transposon vector system and its application in expressing RTPs at high level in mammalian cells are reviewed.
Collapse
Affiliation(s)
- Mian Wei
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang, China
| | - Chun-Liu Mi
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang, China
| | - Chang-Qin Jing
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Chang-Qin Jing, ; Tian-Yun Wang,
| | - Tian-Yun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang, China
- *Correspondence: Chang-Qin Jing, ; Tian-Yun Wang,
| |
Collapse
|
13
|
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
|
14
|
Smirnov A, Battulin N. Concatenation of Transgenic DNA: Random or Orchestrated? Genes (Basel) 2021; 12:genes12121969. [PMID: 34946918 PMCID: PMC8701086 DOI: 10.3390/genes12121969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/18/2022] Open
Abstract
Generation of transgenic organisms by pronuclear microinjection has become a routine procedure. However, while the process of DNA integration in the genome is well understood, we still do not know much about the recombination between transgene molecules that happens in the first moments after DNA injection. Most of the time, injected molecules are joined together in head-to-tail tandem repeats-the so-called concatemers. In this review, we focused on the possible concatenation mechanisms and how they could be studied with genetic reporters tracking individual copies in concatemers. We also discuss various features of concatemers, including palindromic junctions and repeat-induced gene silencing (RIGS). Finally, we speculate how cooperation of DNA repair pathways creates a multicopy concatenated insert.
Collapse
Affiliation(s)
- Alexander Smirnov
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia;
| | - Nariman Battulin
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia;
- Institute of Genetic Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
- Correspondence:
| |
Collapse
|
15
|
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
|
16
|
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
|