51
|
Capella Roca B, Lao N, Barron N, Doolan P, Clynes M. An arginase-based system for selection of transfected CHO cells without the use of toxic chemicals. J Biol Chem 2019; 294:18756-18768. [PMID: 31666335 DOI: 10.1074/jbc.ra119.011162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/29/2019] [Indexed: 11/06/2022] Open
Abstract
Polyamines have essential roles in cell proliferation, DNA replication, transcription, and translation processes, with intracellular depletion of putrescine, spermidine, and spermine resulting in cellular growth arrest and eventual death. Serum-free media for CHO-K1 cells require putrescine supplementation, because these cells lack the first enzyme of the polyamine production pathway, arginase. On the basis of this phenotype, we developed an arginase-based selection system. We transfected CHO-K1 cells with a bicistronic vector co-expressing GFP and arginase and selected cells in media devoid of l-ornithine and putrescine, resulting in mixed populations stably expressing GFP. Moreover, single clones in these selective media stably expressed GFP for a total of 42 generations. Using this polyamine starvation method, we next generated recombinant CHO-K1 cells co-expressing arginase and human erythropoietin (hEPO), which also displayed stable expression and healthy growth. The hEPO-expressing clones grew in commercial media, such as BalanCD and CHO-S serum-free media (SFM)-II, as well as in a defined serum-free, putrescine-containing medium for at least 9 passages (27 generations), with a minimal decrease in hEPO titer by the end of the culture. We observed a lack of arginase activity also in several CHO cell strains (CHO-DP12, CHO-S, and DUXB11) and other mammalian cell lines, including BHK21, suggesting broader utility of this selection system. In conclusion, we have established an easy-to-apply alternative selection system that effectively generates mammalian cell clones expressing biopharmaceutically relevant or other recombinant proteins without the need for any toxic selective agents. We propose that this system is applicable to mammalian cell lines that lack arginase activity.
Collapse
Affiliation(s)
- Berta Capella Roca
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland; SSPC-SFI, Centre for Pharmaceuticals, Dublin City University, Dublin 9, Ireland.
| | - Nga Lao
- National Institute for Bioprocessing Research & Training, A94 X099 Dublin, Ireland
| | - Niall Barron
- National Institute for Bioprocessing Research & Training, A94 X099 Dublin, Ireland; School of Chemical & Bioprocessing Engineering, University College Dublin, Belfield, Dublin 4, D04V1W8, Ireland
| | - Padraig Doolan
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland; SSPC-SFI, Centre for Pharmaceuticals, Dublin City University, Dublin 9, Ireland
| |
Collapse
|
52
|
The Pleiotropic Effects of Glutamine Metabolism in Cancer. Cancers (Basel) 2019; 11:cancers11060770. [PMID: 31167399 PMCID: PMC6627534 DOI: 10.3390/cancers11060770] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/18/2022] Open
Abstract
Metabolic programs are known to be altered in cancers arising from various tissues. Malignant transformation can alter signaling pathways related to metabolism and increase the demand for both energy and biomass for the proliferating cancerous cells. This scenario is further complexed by the crosstalk between transformed cells and the microenvironment. One of the most common metabolic alterations, which occurs in many tissues and in the context of multiple oncogenic drivers, is the increased demand for the amino acid glutamine. Many studies have attributed this increased demand for glutamine to the carbon backbone and its role in the tricarboxylic acid (TCA) cycle anaplerosis. However, an increasing number of studies are now emphasizing the importance of glutamine functioning as a proteogenic building block, a nitrogen donor and carrier, an exchanger for import of other amino acids, and a signaling molecule. Herein, we highlight the recent literature on glutamine’s versatile role in cancer, with a focus on nitrogen metabolism, and therapeutic implications of glutamine metabolism in cancer.
Collapse
|
53
|
Lin PC, Chan KF, Kiess IA, Tan J, Shahreel W, Wong SY, Song Z. Attenuated glutamine synthetase as a selection marker in CHO cells to efficiently isolate highly productive stable cells for the production of antibodies and other biologics. MAbs 2019; 11:965-976. [PMID: 31043114 PMCID: PMC6601560 DOI: 10.1080/19420862.2019.1612690] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are the biopharmaceutical industry’s primary means of manufacturing therapeutic proteins, including monoclonal antibodies. The major challenge in cell line development for the production of recombinant biopharmaceuticals lies in generating and isolating rare high-producing stable clones, amongst thousands of low-producing or unstable clones, in a short period of time. One approach to accomplish this is to use the glutamine synthetase (GS) selection system, together with the GS inhibitor, methionine sulfoximine (MSX). However, MSX can only increase protein productivity to a limited extent. Often productivity will drop when MSX is removed from the system. We evaluated a congenital GS mutation, R324C, which causes glutamine deficiency in human as an attenuated selection marker for CHO cell line generation. We also created a panel of GS mutants with diminished GS activity. Our results demonstrated that using attenuated GS mutants as selection markers significantly increased antibody production of stably transfected pools. Furthermore, these stably transfected pools sustained high productivity levels for an extended period of time, whereas cells transfected with wild-type GS lost considerable protein productivity over time, particularly after MSX was removed. In summary, the use of attenuated GS as a selection marker in CHO cell line development bypasses the need for MSX, and generates stable clones with significantly higher antibody productivity.Abbreviations: CHO: Chinese hamster ovary; CMV: Cytomegalovirus; DHFR: Dihydrofolate reductase; GFP: Green fluorescent protein; GOI: gene-of-interest; GS: Glutamine synthetase; IRES: internal ribosomal entry site; MSX: Methionine sulfoximine; MTX: Methotrexate; psGS: pseudoGS; RVDs: Repeated variable di-residues; TALENs: transcription activator-like effector nucleases; VCD: Viable cell density; ZFNs: zinc finger nucleases.
Collapse
Affiliation(s)
- Pao-Chun Lin
- a Bioprocessing Technology Institute, Agency for Science , Technology and Research (A*STAR) , Singapore
| | - Kah Fai Chan
- a Bioprocessing Technology Institute, Agency for Science , Technology and Research (A*STAR) , Singapore
| | - Irene A Kiess
- a Bioprocessing Technology Institute, Agency for Science , Technology and Research (A*STAR) , Singapore
| | - Joselyn Tan
- a Bioprocessing Technology Institute, Agency for Science , Technology and Research (A*STAR) , Singapore
| | - Wahyu Shahreel
- a Bioprocessing Technology Institute, Agency for Science , Technology and Research (A*STAR) , Singapore
| | - Sze-Yue Wong
- a Bioprocessing Technology Institute, Agency for Science , Technology and Research (A*STAR) , Singapore
| | - Zhiwei Song
- a Bioprocessing Technology Institute, Agency for Science , Technology and Research (A*STAR) , Singapore.,b Department of Biochemistry , Yong Loo Lin School of Medicine, National University of Singapore , Singapore
| |
Collapse
|
54
|
Ong EC, Smidt P, McGrew JT. Limiting the metabolic burden of recombinant protein expression during selection yields pools with higher expression levels. Biotechnol Prog 2019; 35:e2839. [PMID: 31090257 DOI: 10.1002/btpr.2839] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/02/2019] [Accepted: 05/13/2019] [Indexed: 12/13/2022]
Abstract
In order to avoid the metabolic burden of protein expression during cell growth, and to avoid potential toxicity of recombinant proteins, microbial expression systems typically utilize regulated expression vectors. In contrast, constitutive expression vectors have usually been utilized for isolation of protein expressing mammalian cell lines. In mammalian systems, inducible expression vectors are typically utilized for only those proteins that are toxic when overexpressed. We developed a tetracycline regulated expression system in CHO cells, and show that cell pools selected in the uninduced state recover faster than those selected in the induced state even though the proteins showed no apparent toxicity or expression instability. Furthermore, cell pools selected in the uninduced state had higher expression levels when protein expression was turned on only in production cultures compared to pools that were selected and maintained in the induced state through production. We show a titer improvement of greater than twofold for an Fc-fusion protein and greater than 50% improvement for a recombinant antibody. The improvement is primarily due to an increase in specific productivity. Recombinant protein mRNA levels correlate strongly with protein expression levels and are highest in those cultures selected in the uninduced state and only induced during production. These data are consistent with a model where CHO cell lines with constitutive expression select for subclones with lower expression levels.
Collapse
Affiliation(s)
- E-Ching Ong
- Process Design, Just Biotherapeutics, Seattle, Washington
| | - Pauline Smidt
- Process Design, Just Biotherapeutics, Seattle, Washington
| | | |
Collapse
|
55
|
Zhu J, Hatton D. New Mammalian Expression Systems. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 165:9-50. [PMID: 28585079 DOI: 10.1007/10_2016_55] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There are an increasing number of recombinant antibodies and proteins in preclinical and clinical development for therapeutic applications. Mammalian expression systems are key to enabling the production of these molecules, and Chinese hamster ovary (CHO) cell platforms continue to be central to delivery of the stable cell lines required for large-scale production. Increasing pressure on timelines and efficiency, further innovation of molecular formats and the shift to new production systems are driving developments of these CHO cell line platforms. The availability of genome and transcriptome data coupled with advancing gene editing tools are increasing the ability to design and engineer CHO cell lines to meet these challenges. This chapter aims to give an overview of the developments in CHO expression systems and some of the associated technologies over the past few years.
Collapse
Affiliation(s)
- Jie Zhu
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Diane Hatton
- MedImmune, Milstein Building, Granta Park, Cambridge, CB21 6GH, UK.
| |
Collapse
|
56
|
Yang B, Zhou J, Zhao H, Wang A, Lei Y, Xie Q, Xiong S. Study of the mechanism for increased protein expression via transcription potency reduction of the selection marker. Bioprocess Biosyst Eng 2019; 42:799-806. [PMID: 30730009 DOI: 10.1007/s00449-019-02083-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/24/2019] [Indexed: 01/02/2023]
Abstract
Stable transfection of mammalian cells using various expression cassettes for exogenous gene expression has been well established. The impact of critical factors in these cassettes, such as promoter and enhancer elements, on recombinant protein production in mammalian cells has been studied extensively to optimize the expression efficiency. However, few studies on the correlation between the strength of selection marker and the expression of gene of interest (GOI) have been reported. Here we investigated the correlation between the strength of a widely used selection marker, glutamine synthetase (GS) gene, and gene of interest in which the expression of GOI is driven by mouse cytomegalovirus (mCMV) major immediate early (MIE) promoter whereas the expression of GS is controlled by SV40E (Simian vacuolating virus 40E) promoter. We used a green fluorescent protein and the adalimumab antibody (heavy and light chain) as two distinct examples for the gene of interest. We then decreased the expression of GS gene by engineering a specific region of its SV40E promoter in these expression cassettes. By comparing the expression of GS and GOI at transcription and translation level before and after the SV40E promoter was weakened, we found that lower GS expression due to weaker SV40E transcription correlated well with the higher expression of recombinant proteins, mainly by increasing the copy number of GS and GOI integration into host cell genome.
Collapse
Affiliation(s)
- Bin Yang
- Department of Cell Biol, Institute of Biomedicine & National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, 730 Building of Biology, 601 W Huangpu Ave, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Jiatao Zhou
- Sunshine Lake Pharma Co., Ltd., Zhen An Road, Dongguan, 523867, People's Republic of China
| | - Hui Zhao
- Department of Cell Biol, Institute of Biomedicine & National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, 730 Building of Biology, 601 W Huangpu Ave, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Anling Wang
- Department of Cell Biol, Institute of Biomedicine & National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, 730 Building of Biology, 601 W Huangpu Ave, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Yuanjun Lei
- Department of Cell Biol, Institute of Biomedicine & National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, 730 Building of Biology, 601 W Huangpu Ave, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Qiuling Xie
- Department of Cell Biol, Institute of Biomedicine & National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, 730 Building of Biology, 601 W Huangpu Ave, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Sheng Xiong
- Department of Cell Biol, Institute of Biomedicine & National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, 730 Building of Biology, 601 W Huangpu Ave, Guangzhou, 510630, Guangdong, People's Republic of China.
| |
Collapse
|
57
|
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]
|
58
|
|
59
|
Hong JK, Lakshmanan M, Goudar C, Lee DY. Towards next generation CHO cell line development and engineering by systems approaches. Curr Opin Chem Eng 2018. [DOI: 10.1016/j.coche.2018.08.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
60
|
Hamaker NK, Lee KH. Site-specific Integration Ushers in a New Era of Precise CHO Cell Line Engineering. Curr Opin Chem Eng 2018; 22:152-160. [PMID: 31086757 DOI: 10.1016/j.coche.2018.09.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chinese hamster ovary (CHO) cells are widely used for the production of therapeutic proteins. Customarily, CHO production cell lines are established through random integration, which requires laborious screening of many clones to isolate suitable producers. In contrast, site-specific integration (SSI) accelerates cell line development by targeting integration of transgenes to pre-validated genomic loci capable of supporting high and stable expression. To date, a relatively small number of these so called 'hot spots' have been identified, mainly through empirical methods. Nevertheless, nuclease-mediated and recombinase-mediated SSI have revolutionized cell line engineering by enabling rational and reproducible transgene targeting.
Collapse
Affiliation(s)
- Nathaniel K Hamaker
- Delaware Biotechnology Institute, Newark, DE.,Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Kelvin H Lee
- Delaware Biotechnology Institute, Newark, DE.,Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| |
Collapse
|
61
|
Ritacco FV, Wu Y, Khetan A. Cell culture media for recombinant protein expression in Chinese hamster ovary (CHO) cells: History, key components, and optimization strategies. Biotechnol Prog 2018; 34:1407-1426. [DOI: 10.1002/btpr.2706] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Frank V. Ritacco
- Biologics Process DevelopmentBristol‐Myers Squibb Pennington New Jersey United States
| | - Yongqi Wu
- Biologics Process DevelopmentBristol‐Myers Squibb Pennington New Jersey United States
| | - Anurag Khetan
- Biologics Process DevelopmentBristol‐Myers Squibb Pennington New Jersey United States
| |
Collapse
|
62
|
Hunter M, Yuan P, Vavilala D, Fox M. Optimization of Protein Expression in Mammalian Cells. ACTA ACUST UNITED AC 2018; 95:e77. [DOI: 10.1002/cpps.77] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
63
|
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
|
64
|
Yang Y, You M, Chen F, Jia T, Chen Y, Zhou B, Mi Q, An Z, Luo W, Xia N. Efficient development of a stable cell pool for antibody production using a single plasmid. J Biochem 2018; 163:391-398. [PMID: 29361116 DOI: 10.1093/jb/mvy007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 11/07/2017] [Indexed: 01/02/2023] Open
Abstract
Therapeutic antibodies are the fastest growing group of biopharmaceuticals. Evaluation of drug candidates requires a sufficient amount of antibodies. Production of antibodies with stable cell pools is an efficient strategy to produce grams of proteins for drug candidate selection. Many methods have been described for developing stable cell pools for antibody expression. However, most of the reported methods are laborious due to the low frequency of high producers. In this study, we determined optimal vectors and screening parameters to develop a strategy for efficient construction of stable antibody expressing cell pools. The cell pool constructed using the optimized strategy consistently yielded a higher expression titer, up to 10-fold improvement. Further, this method resulted in a higher ratio of the cell pools with the main product peak above 95% as assessed by size-exclusion chromatography. High producers could be obtained by means of screening five 96-well plates. This strategy will greatly reduce clone-screening size during Clinical Lead Selection. This study provides a platform with efficient design of plasmids and screening strategies for significant cost and labour savings in high expression of two-subunit proteins such as antibodies.
Collapse
Affiliation(s)
- Yi Yang
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen 361102, People's Republic of China.,State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China
| | - Min You
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen 361102, People's Republic of China.,State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China
| | - Fentian Chen
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen 361102, People's Republic of China.,State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China
| | - Tianrong Jia
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen 361102, People's Republic of China.,State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China
| | - Yuanzhi Chen
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen 361102, People's Republic of China.,State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China
| | - Bing Zhou
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen 361102, People's Republic of China.,State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China
| | - Qingyu Mi
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen 361102, People's Republic of China.,State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China
| | - Zhiqiang An
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China.,Texas Therapeutics Institute, The Brown Foundation of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wenxin Luo
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen 361102, People's Republic of China.,State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China
| | - Ningshao Xia
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen 361102, People's Republic of China.,State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen 361102, People's Republic of China
| |
Collapse
|
65
|
Noh SM, Shin S, Lee GM. Comprehensive characterization of glutamine synthetase-mediated selection for the establishment of recombinant CHO cells producing monoclonal antibodies. Sci Rep 2018; 8:5361. [PMID: 29599455 PMCID: PMC5876325 DOI: 10.1038/s41598-018-23720-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/18/2018] [Indexed: 12/16/2022] Open
Abstract
To characterize a glutamine synthetase (GS)-based selection system, monoclonal antibody (mAb) producing recombinant CHO cell clones were generated by a single round of selection at various methionine sulfoximine (MSX) concentrations (0, 25, and 50 μM) using two different host cell lines (CHO-K1 and GS-knockout CHO). Regardless of the host cell lines used, the clones selected at 50 μM MSX had the lowest average specific growth rate and the highest average specific production rates of toxic metabolic wastes, lactate and ammonia. Unlike CHO-K1, high producing clones could be generated in the absence of MSX using GS-knockout CHO with an improved selection stringency. Regardless of the host cell lines used, the clones selected at various MSX concentrations showed no significant difference in the GS, heavy chain, and light chain gene copies (P > 0.05). Furthermore, there was no correlation between the specific mAb productivity and these three gene copies (R2 ≤ 0.012). Taken together, GS-mediated gene amplification does not occur in a single round of selection at a MSX concentration up to 50 μM. The use of the GS-knockout CHO host cell line facilitates the rapid generation of high producing clones with reduced production of lactate and ammonia in the absence of MSX.
Collapse
Affiliation(s)
- Soo Min Noh
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seunghyeon Shin
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea. .,The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Hørsholm, Denmark.
| |
Collapse
|
66
|
Handlogten MW, Lee-O'Brien A, Roy G, Levitskaya SV, Venkat R, Singh S, Ahuja S. Intracellular response to process optimization and impact on productivity and product aggregates for a high-titer CHO cell process. Biotechnol Bioeng 2017; 115:126-138. [DOI: 10.1002/bit.26460] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 08/19/2017] [Accepted: 09/19/2017] [Indexed: 12/24/2022]
Affiliation(s)
| | - Allison Lee-O'Brien
- Cell Culture and Fermentation Sciences; MedImmune LLC; Gaithersburg Maryland
| | - Gargi Roy
- Antibody Discovery and Protein Engineering; MedImmune LLC; Gaithersburg Maryland
| | | | - Raghavan Venkat
- Cell Culture and Fermentation Sciences; MedImmune LLC; Gaithersburg Maryland
| | - Shailendra Singh
- Biologics Development and Commercialization; West Point Pennsylvania
| | - Sanjeev Ahuja
- Cell Culture and Fermentation Sciences; MedImmune LLC; Gaithersburg Maryland
| |
Collapse
|
67
|
Yeo JHM, Ho SCL, Mariati M, Koh E, Tay SJ, Woen S, Zhang P, Yang Y. Optimized Selection Marker and CHO Host Cell Combinations for Generating High Monoclonal Antibody Producing Cell Lines. Biotechnol J 2017; 12. [DOI: 10.1002/biot.201700175] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 09/13/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Jessna H. M. Yeo
- Bioprocessing Technology Institute; Agency for Science; Technology and Research (A*STAR) Singapore Singapore
| | - Steven C. L. Ho
- Bioprocessing Technology Institute; Agency for Science; Technology and Research (A*STAR) Singapore Singapore
| | - Mariati Mariati
- Bioprocessing Technology Institute; Agency for Science; Technology and Research (A*STAR) Singapore Singapore
| | - Esther Koh
- Bioprocessing Technology Institute; Agency for Science; Technology and Research (A*STAR) Singapore Singapore
| | - Shi Jie Tay
- Bioprocessing Technology Institute; Agency for Science; Technology and Research (A*STAR) Singapore Singapore
| | - Susanto Woen
- Bioprocessing Technology Institute; Agency for Science; Technology and Research (A*STAR) Singapore Singapore
| | - Peiqing Zhang
- Bioprocessing Technology Institute; Agency for Science; Technology and Research (A*STAR) Singapore Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute; Agency for Science; Technology and Research (A*STAR) Singapore Singapore
| |
Collapse
|
68
|
Wright C, Alves C, Kshirsagar R, Pieracci J, Estes S. Leveraging a CHO cell line toolkit to accelerate biotherapeutics into the clinic. Biotechnol Prog 2017; 33:1468-1475. [DOI: 10.1002/btpr.2548] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/03/2017] [Indexed: 01/29/2023]
Affiliation(s)
| | | | | | | | - Scott Estes
- Codiak Biosciences, Upstream Process Development; Cambridge MA
| |
Collapse
|
69
|
Brown AJ, Kalsi D, Fernandez-Martell A, Cartwright J, Barber NOW, Patel YD, Turner R, Bryant CL, Johari YB, James DC. Expression Systems for Recombinant Biopharmaceutical Production by Mammalian Cells in Culture. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1002/9783527699124.ch13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Adam J. Brown
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Devika Kalsi
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | | | - Joe Cartwright
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Nicholas O. W. Barber
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Yash D. Patel
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | | | - Claire L. Bryant
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Yusuf B. Johari
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - David C. James
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| |
Collapse
|
70
|
Poulain A, Perret S, Malenfant F, Mullick A, Massie B, Durocher Y. Rapid protein production from stable CHO cell pools using plasmid vector and the cumate gene-switch. J Biotechnol 2017. [PMID: 28625678 DOI: 10.1016/j.jbiotec.2017.06.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
To rapidly produce large amounts of recombinant proteins, the generation of stable Chinese Hamster Ovary (CHO) cell pools represents a useful alternative to large-scale transient gene expression (TGE). We have developed a cell line (CHOBRI/rcTA) allowing the inducible expression of recombinant proteins, based on the cumate gene switch. After the identification of optimal plasmid DNA topology (supercoiled vs linearized plasmid) for PEIpro™ mediated transfection and of optimal conditions for methionine sulfoximine (MSX) selection, we were able to generate CHOBRI/rcTA pools producing high levels of recombinant proteins. Volumetric productivities of up to 900mg/L were reproducibly achieved for a Fc fusion protein and up to 350mg/L for an antibody after 14days post-induction in non-optimized fed-batch cultures. In addition, we show that CHO pool volumetric productivities are not affected by a freeze-thaw cycle or following maintenance in culture for over one month in the presence of MSX. Finally, we demonstrate that volumetric protein production with the CR5 cumate-inducible promoter is three- to four-fold higher than with the human CMV or hybrid EF1α-HTLV constitutive promoters. These results suggest that the cumate-inducible CHOBRI/rcTA stable pool platform is a powerful and robust system for the rapid production of gram amounts of recombinant proteins.
Collapse
Affiliation(s)
- Adeline Poulain
- National Research Council of Canada, 6100 Royalmount Avenue, Montréal, QC H4P 2R2, Canada; Département de Microbiologie et Immunologie, Faculté de Médecine, Université de Montréal, QC, Canada
| | - Sylvie Perret
- National Research Council of Canada, 6100 Royalmount Avenue, Montréal, QC H4P 2R2, Canada
| | - Félix Malenfant
- National Research Council of Canada, 6100 Royalmount Avenue, Montréal, QC H4P 2R2, Canada
| | - Alaka Mullick
- National Research Council of Canada, 6100 Royalmount Avenue, Montréal, QC H4P 2R2, Canada
| | - Bernard Massie
- National Research Council of Canada, 6100 Royalmount Avenue, Montréal, QC H4P 2R2, Canada; Département de Microbiologie et Immunologie, Faculté de Médecine, Université de Montréal, QC, Canada
| | - Yves Durocher
- National Research Council of Canada, 6100 Royalmount Avenue, Montréal, QC H4P 2R2, Canada; Département de Biochimie et Médecine Moléculaire, Faculté de Médecine, Université de Montréal, QC, Canada.
| |
Collapse
|
71
|
Richelle A, Lewis NE. Improvements in protein production in mammalian cells from targeted metabolic engineering. ACTA ACUST UNITED AC 2017; 6:1-6. [PMID: 29104947 DOI: 10.1016/j.coisb.2017.05.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bioprocess optimization has yielded powerful clones for biotherapeutic production. However, new genomic technologies allow more targeted approaches to cell line development. Here we review efforts to enhance protein production in mammalian cells through metabolic engineering. Most efforts aimed to reduce toxic byproducts accumulation to enhance protein productivity. However, recent work highlights the possibility of regulating other desirable traits (e.g., apoptosis and glycosylation) by targeting central metabolism since these processes are interconnected. Therefore, as we further detail the pathways underlying cell growth and protein production and deploy diverse algorithms for their analysis, opportunities will arise to move beyond simple cell line designs and facilitate cell engineering strategies with complex combinations of genes that together underlie a phenotype of interest.
Collapse
Affiliation(s)
- Anne Richelle
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States.,Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States
| | - Nathan E Lewis
- Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States.,Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States
| |
Collapse
|
72
|
Xu N, Ma C, Ou J, Sun WW, Zhou L, Hu H, Liu XM. Comparative Proteomic Analysis of Three Chinese Hamster Ovary (CHO) Host Cells. Biochem Eng J 2017; 124:122-129. [PMID: 28736500 DOI: 10.1016/j.bej.2017.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chinese hamster ovary (CHO) cells have been widely used to express heterologous genes and produce therapeutic proteins in biopharmaceutical industry. Different CHO host cells have distinct cell growth rates and protein expression characteristics. In this study, the expression of about 1,307 host proteins in three sublines, i.e. CHO K1, CHO S and CHO/dihydrofolate reductase (dhfr)- , were investigated and compared using proteomic analysis. The proteins involved in cell growth, glycolysis, tricarboxylic acid cycle, transcription, translation and glycosylation were quantitated using Liquid chromatography tandem-mass spectrometry (LC-MS/MS). The key host cell proteins that regulate the kinetics of cell growth and the magnitude of protein expression levels were identified. Furthermore, several rational cell engineering strategies on how to combine the desired features of fast cell growth and efficient production of therapeutic proteins into one new super CHO host cell have been proposed.
Collapse
Affiliation(s)
- Ningning Xu
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1670 University Blvd, Birmingham, AL 35233, USA
| | - Chao Ma
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1670 University Blvd, Birmingham, AL 35233, USA
| | - Jianfa Ou
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1670 University Blvd, Birmingham, AL 35233, USA
| | - Wanqi Wendy Sun
- Department of Chemical and Biological Engineering, The University of Alabama (UA), 245 7th Avenue, Tuscaloosa, AL 35401, USA
| | - Lufang Zhou
- Departments of Medicine and Biomedical Engineering, University of Alabama at Birmingham (UAB), 703 19 Street South and 1530 3 Avenue South, Birmingham, AL 35294, USA
| | - Hui Hu
- Department of Microbiology, University of Alabama at Birmingham (UAB), 845 19 Street South, Birmingham AL 35294, USA
| | - Xiaoguang Margaret Liu
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1670 University Blvd, Birmingham, AL 35233, USA
| |
Collapse
|
73
|
Fan L, Rizzi G, Bierilo K, Tian J, Yee JC, Russell R, Das TK. Comparative study of therapeutic antibody candidates derived from mini-pool and clonal cell lines. Biotechnol Prog 2017; 33:1456-1462. [DOI: 10.1002/btpr.2477] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/03/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Lianchun Fan
- Cellular Molecular Biology; Bristol-Myers Squibb Company; Pennington NJ 08534
| | - Giovanni Rizzi
- Cellular Molecular Biology; Bristol-Myers Squibb Company; Pennington NJ 08534
| | - Kathleen Bierilo
- Cellular Molecular Biology; Bristol-Myers Squibb Company; Pennington NJ 08534
| | - Jun Tian
- Process Development; Bristol-Myers Squibb Company; Devens MA 01434
| | - Joon Chong Yee
- Process Development; Bristol-Myers Squibb Company; Devens MA 01434
| | - Reb Russell
- Analytical and Process Development; Bristol-Myers Squibb Company; Pennington NJ 08534
| | - Tapan K Das
- Biologics Characterization & Analytical Development; Bristol-Myers Squibb Company; Pennington NJ 08534
| |
Collapse
|
74
|
Rajendra Y, Balasubramanian S, Peery RB, Swartling JR, McCracken NA, Norris DL, Frye CC, Barnard GC. Bioreactor scale up and protein product quality characterization of piggyBac transposon derived CHO pools. Biotechnol Prog 2017; 33:534-540. [PMID: 28188692 DOI: 10.1002/btpr.2447] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/06/2017] [Indexed: 11/10/2022]
Abstract
Chinese hamster ovary (CHO) cells remain the most popular host for the production of biopharmaceutical drugs, particularly monoclonal antibodies (mAbs), bispecific antibodies, and Fc-fusion proteins. Creating and characterizing the stable CHO clonally-derived cell lines (CDCLs) needed to manufacture these therapeutic proteins is a lengthy and laborious process. Therefore, CHO pools have increasingly been used to rapidly produce protein to support and enable preclinical drug development. We recently described the generation of CHO pools yielding mAb titers as high as 7.6 g/L in a 16 day bioprocess using piggyBac transposon-mediated gene integration. In this study, we wanted to understand why the piggyBac pool titers were significantly higher (2-10 fold) than the control CHO pools. Higher titers were the result of a combination of increased average gene copy number, significantly higher messenger RNA levels and the homogeneity (i.e. less diverse population distribution) of the piggyBac pools, relative to the control pools. In order to validate the use of piggyBac pools to support preclinical drug development, we then performed an in-depth product quality analysis of purified protein. The product quality of protein obtained from the piggyBac pools was very similar to the product quality profile of protein obtained from the control pools. Finally, we demonstrated the scalability of these pools from shake flasks to 36L bioreactors. Overall, these results suggest that gram quantities of therapeutic protein can be rapidly obtained from piggyBac CHO pools without significantly changing product quality attributes. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:534-540, 2017.
Collapse
Affiliation(s)
- Yashas Rajendra
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46225
| | - Sowmya Balasubramanian
- Bioprocess Research and Development, Eli Lilly and Company, LTC-North, 1200 Kentucky Avenue, Indianapolis, IN, 46221
| | - Robert B Peery
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46225
| | - James R Swartling
- Bioprocess Research and Development, Eli Lilly and Company, LTC-North, 1200 Kentucky Avenue, Indianapolis, IN, 46221
| | - Neil A McCracken
- Bioprocess Research and Development, Eli Lilly and Company, LTC-North, 1200 Kentucky Avenue, Indianapolis, IN, 46221
| | - Dawn L Norris
- Bioprocess Research and Development, Eli Lilly and Company, LTC-North, 1200 Kentucky Avenue, Indianapolis, IN, 46221
| | - Christopher C Frye
- Bioprocess Research and Development, Eli Lilly and Company, LTC-North, 1200 Kentucky Avenue, Indianapolis, IN, 46221
| | - Gavin C Barnard
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46225
| |
Collapse
|
75
|
Hsu HH, Araki M, Mochizuki M, Hori Y, Murata M, Kahar P, Yoshida T, Hasunuma T, Kondo A. A Systematic Approach to Time-series Metabolite Profiling and RNA-seq Analysis of Chinese Hamster Ovary Cell Culture. Sci Rep 2017; 7:43518. [PMID: 28252038 PMCID: PMC5333161 DOI: 10.1038/srep43518] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/27/2017] [Indexed: 11/11/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are the primary host used for biopharmaceutical protein production. The engineering of CHO cells to produce higher amounts of biopharmaceuticals has been highly dependent on empirical approaches, but recent high-throughput "omics" methods are changing the situation in a rational manner. Omics data analyses using gene expression or metabolite profiling make it possible to identify key genes and metabolites in antibody production. Systematic omics approaches using different types of time-series data are expected to further enhance understanding of cellular behaviours and molecular networks for rational design of CHO cells. This study developed a systematic method for obtaining and analysing time-dependent intracellular and extracellular metabolite profiles, RNA-seq data (enzymatic mRNA levels) and cell counts from CHO cell cultures to capture an overall view of the CHO central metabolic pathway (CMP). We then calculated correlation coefficients among all the profiles and visualised the whole CMP by heatmap analysis and metabolic pathway mapping, to classify genes and metabolites together. This approach provides an efficient platform to identify key genes and metabolites in CHO cell culture.
Collapse
Affiliation(s)
- Han-Hsiu Hsu
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Michihiro Araki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Masao Mochizuki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Yoshimi Hori
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Masahiro Murata
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Prihardi Kahar
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Takanobu Yoshida
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Tomohisa Hasunuma
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Akihiko Kondo
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| |
Collapse
|
76
|
Rajendra Y, Peery RB, Hougland MD, Barnard GC, Wu X, Fitchett JR, Bacica M, Demarest SJ. Transient and stable CHO expression, purification and characterization of novel hetero-dimeric bispecific IgG antibodies. Biotechnol Prog 2017; 33:469-477. [PMID: 27977915 DOI: 10.1002/btpr.2414] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/03/2016] [Indexed: 01/05/2023]
Abstract
IgG bispecific antibodies (BsAbs) represent one of the preferred formats for bispecific antibody therapeutics due to their native-like IgG properties and their monovalent binding to each target. Most reported studies utilized transient expression in HEK293 cells to produce BsAbs. However, the expression of biotherapeutic molecules using stable CHO cell lines is commonly used for biopharmaceutical manufacturing. Unfortunately, limited information is available in the scientific literature on the expression of BsAbs in CHO cell lines. In this study we describe an alternative approach to express the multiple components of IgG BsAbs using a single plasmid vector (quad vector). This single plasmid vector contains both heavy chain genes and both light chain genes required for the expression and assembly of the IgG BsAb, along with a selectable marker. We expressed, purified, and characterized four different IgG BsAbs or "hetero-mAbs" using transient CHO expression and stable CHO minipools. Transient CHO titers ranged from 90 to 160 mg/L. Stable CHO titers ranged from 0.4 to 2.3 g/L. Following a simple Protein A purification step, the percentage of correctly paired BsAbs ranged from 74% to 98% as determined by mass spectrometry. We also found that information generated from transient CHO expression was similar to information generated using stable CHO minipools. In conclusion, the quad vector approach represents a simple, but effective, alternative approach for the generation of IgG BsAbs in both transient CHO and stable CHO expression systems. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:469-477, 2017.
Collapse
Affiliation(s)
- Yashas Rajendra
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285
| | - Robert B Peery
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285
| | - Maria D Hougland
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285
| | - Gavin C Barnard
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285
| | - Xiufeng Wu
- Biotechnology Discovery Research, Lilly Biotechnology Center, San Diego, CA, 92121
| | - Jonathan R Fitchett
- Biotechnology Discovery Research, Lilly Biotechnology Center, San Diego, CA, 92121
| | - Michael Bacica
- Biotechnology Discovery Research, Lilly Biotechnology Center, San Diego, CA, 92121
| | - Stephen J Demarest
- Biotechnology Discovery Research, Lilly Biotechnology Center, San Diego, CA, 92121
| |
Collapse
|
77
|
Grav LM, la Cour Karottki KJ, Lee JS, Kildegaard HF. Application of CRISPR/Cas9 Genome Editing to Improve Recombinant Protein Production in CHO Cells. Methods Mol Biol 2017; 1603:101-118. [PMID: 28493126 DOI: 10.1007/978-1-4939-6972-2_7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Genome editing has become an increasingly important aspect of Chinese Hamster Ovary (CHO ) cell line engineering for improving production of recombinant protein therapeutics. Currently, the focus is directed toward expanding the product diversity, controlling and improving product quality and yields. In this chapter, we present our protocol on how to use the genome editing tool Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) to knockout engineering target genes in CHO cells. As an example, we refer to the glutamine synthetase (GS)-encoding gene as the knockout target gene, a knockout that increases the selection efficiency of the GS-mediated gene amplification system.
Collapse
Affiliation(s)
- Lise Marie Grav
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Karen Julie la Cour Karottki
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Jae Seong Lee
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Helene Faustrup Kildegaard
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
78
|
mTORC1 signalling and eIF4E/4E-BP1 translation initiation factor stoichiometry influence recombinant protein productivity from GS-CHOK1 cells. Biochem J 2016; 473:4651-4664. [PMID: 27760840 PMCID: PMC5147049 DOI: 10.1042/bcj20160845] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/17/2016] [Accepted: 10/19/2016] [Indexed: 12/14/2022]
Abstract
Many protein-based biotherapeutics are produced in cultured Chinese hamster ovary (CHO) cell lines. Recent reports have demonstrated that translation of recombinant mRNAs and global control of the translation machinery via mammalian target of rapamycin (mTOR) signalling are important determinants of the amount and quality of recombinant protein such cells can produce. mTOR complex 1 (mTORC1) is a master regulator of cell growth/division, ribosome biogenesis and protein synthesis, but the relationship between mTORC1 signalling, cell growth and proliferation and recombinant protein yields from mammalian cells, and whether this master regulating signalling pathway can be manipulated to enhance cell biomass and recombinant protein production (rPP) are not well explored. We have investigated mTORC1 signalling and activity throughout batch culture of a panel of sister recombinant glutamine synthetase-CHO cell lines expressing different amounts of a model monoclonal IgG4, to evaluate the links between mTORC1 signalling and cell proliferation, autophagy, recombinant protein expression, global protein synthesis and mRNA translation initiation. We find that the expression of the mTORC1 substrate 4E-binding protein 1 (4E-BP1) fluctuates throughout the course of cell culture and, as expected, that the 4E-BP1 phosphorylation profiles change across the culture. Importantly, we find that the eIF4E/4E-BP1 stoichiometry positively correlates with cell productivity. Furthermore, eIF4E amounts appear to be co-regulated with 4E-BP1 amounts. This may reflect a sensing of either change at the mRNA level as opposed to the protein level or the fact that the phosphorylation status, as well as the amount of 4E-BP1 present, is important in the co-regulation of eIF4E and 4E-BP1.
Collapse
|
79
|
Rajendra Y, Peery RB, Barnard GC. Generation of stable Chinese hamster ovary pools yielding antibody titers of up to 7.6 g/L using the piggyBac transposon system. Biotechnol Prog 2016; 32:1301-1307. [DOI: 10.1002/btpr.2307] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/25/2016] [Indexed: 01/16/2023]
Affiliation(s)
- Yashas Rajendra
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center; Indianapolis IN 46285
| | - Robert B. Peery
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center; Indianapolis IN 46285
| | - Gavin C. Barnard
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center; Indianapolis IN 46285
| |
Collapse
|
80
|
Priola JJ, Calzadilla N, Baumann M, Borth N, Tate CG, Betenbaugh MJ. High-throughput screening and selection of mammalian cells for enhanced protein production. Biotechnol J 2016; 11:853-65. [DOI: 10.1002/biot.201500579] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 02/09/2016] [Accepted: 05/17/2016] [Indexed: 01/05/2023]
Affiliation(s)
- Joseph J. Priola
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; Baltimore MD USA
| | - Nathan Calzadilla
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; Baltimore MD USA
| | | | - Nicole Borth
- Department of Biotechnology; Universität für Bodenkultur; Vienna Austria
| | | | - Michael J. Betenbaugh
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; Baltimore MD USA
| |
Collapse
|
81
|
McVey D, Aronov M, Rizzi G, Cowan A, Scott C, Megill J, Russell R, Tirosh B. CHO cells knocked out for TSC2 display an improved productivity of antibodies under fed batch conditions. Biotechnol Bioeng 2016; 113:1942-52. [DOI: 10.1002/bit.25951] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Duncan McVey
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - Michael Aronov
- Institute for Drug Research; The School of Pharmacy; The Hebrew University of Jerusalem; Jerusalem 91120 Israel
| | - Giovanni Rizzi
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - Alexis Cowan
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - Charo Scott
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - John Megill
- Discovery Toxicology; Bristol Myers Squibb Company; Pennington New Jersey
| | - Reb Russell
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
| | - Boaz Tirosh
- Division of Global Manufacturing and Supply; Bristol Myers Squibb Company; Bloomsbury New Jersey
- Institute for Drug Research; The School of Pharmacy; The Hebrew University of Jerusalem; Jerusalem 91120 Israel
| |
Collapse
|
82
|
Villaverde AF, Bongard S, Mauch K, Balsa-Canto E, Banga JR. Metabolic engineering with multi-objective optimization of kinetic models. J Biotechnol 2016; 222:1-8. [PMID: 26826510 DOI: 10.1016/j.jbiotec.2016.01.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 12/30/2015] [Accepted: 01/11/2016] [Indexed: 10/22/2022]
Abstract
Kinetic models have a great potential for metabolic engineering applications. They can be used for testing which genetic and regulatory modifications can increase the production of metabolites of interest, while simultaneously monitoring other key functions of the host organism. This work presents a methodology for increasing productivity in biotechnological processes exploiting dynamic models. It uses multi-objective dynamic optimization to identify the combination of targets (enzymatic modifications) and the degree of up- or down-regulation that must be performed in order to optimize a set of pre-defined performance metrics subject to process constraints. The capabilities of the approach are demonstrated on a realistic and computationally challenging application: a large-scale metabolic model of Chinese Hamster Ovary cells (CHO), which are used for antibody production in a fed-batch process. The proposed methodology manages to provide a sustained and robust growth in CHO cells, increasing productivity while simultaneously increasing biomass production, product titer, and keeping the concentrations of lactate and ammonia at low values. The approach presented here can be used for optimizing metabolic models by finding the best combination of targets and their optimal level of up/down-regulation. Furthermore, it can accommodate additional trade-offs and constraints with great flexibility.
Collapse
Affiliation(s)
- Alejandro F Villaverde
- Bioprocess Engineering Group, IIM-CSIC, Eduardo Cabello 6, 36208 Vigo, Spain; Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; Department of Systems and Control Engineering, Universidade de Vigo, Rua Maxwell, 36310 Vigo, Spain
| | - Sophia Bongard
- Insilico Biotechnology AG, Meitnerstraße 9, 70563 Stuttgart, Germany
| | - Klaus Mauch
- Insilico Biotechnology AG, Meitnerstraße 9, 70563 Stuttgart, Germany
| | - Eva Balsa-Canto
- Bioprocess Engineering Group, IIM-CSIC, Eduardo Cabello 6, 36208 Vigo, Spain
| | - Julio R Banga
- Bioprocess Engineering Group, IIM-CSIC, Eduardo Cabello 6, 36208 Vigo, Spain
| |
Collapse
|
83
|
Lian Z, Wu Q, Wang T. Identification and characterization of a -1 reading frameshift in the heavy chain constant region of an IgG1 recombinant monoclonal antibody produced in CHO cells. MAbs 2015; 8:358-70. [PMID: 26652198 PMCID: PMC4966638 DOI: 10.1080/19420862.2015.1116658] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/27/2015] [Accepted: 11/01/2015] [Indexed: 01/16/2023] Open
Abstract
Frameshifts lead to complete alteration of the intended amino acid sequences, and therefore may affect the biological activities of protein therapeutics and pose potential immunogenicity risks. We report here the identification and characterization of a novel -1 frameshift variant in a recombinant IgG1 therapeutic monoclonal antibody (mAb) produced in Chinese hamster ovary cells during the cell line selection studies. The variant was initially observed as an atypical post-monomer fragment peak in size exclusion chromatography. Characterization of the fragment peak using intact and reduced liquid chromatography-mass spectrometry (LC-MS) analyses determined that the fragment consisted of a normal light chain disulfide-linked to an aberrant 26 kDa fragment that could not be assigned to any HC fragment even after considering common modifications. Further analysis using LC-MS/MS peptide mapping revealed that the aberrant fragment contained the expected HC amino acid sequence (1-232) followed by a 20-mer novel sequence corresponding to expression of heavy chain DNA sequence in the -1 reading frame. Examination of the DNA sequence around the frameshift initiation site revealed that a mononucleotide repeat GGGGGG located in the IgG1 HC constant region was most likely the structural root cause of the frameshift. Rapid identification of the frameshift allowed us to avoid use of a problematic cell line containing the frameshift as the production cell line. The frameshift reported here may be observed in other mAb products and the hypothesis-driven analytical approaches employed here may be valuable for rapid identification and characterization of frameshift variants in other recombinant proteins.
Collapse
Affiliation(s)
- Zhirui Lian
- Bioproduct Research and Development, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Qindong Wu
- Bioproduct Research and Development, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Tongtong Wang
- Bioproduct Research and Development, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| |
Collapse
|
84
|
The art of CHO cell engineering: A comprehensive retrospect and future perspectives. Biotechnol Adv 2015; 33:1878-96. [DOI: 10.1016/j.biotechadv.2015.10.015] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/21/2015] [Accepted: 10/30/2015] [Indexed: 12/14/2022]
|
85
|
Chowdhury R, Chowdhury A, Maranas CD. Using Gene Essentiality and Synthetic Lethality Information to Correct Yeast and CHO Cell Genome-Scale Models. Metabolites 2015; 5:536-70. [PMID: 26426067 PMCID: PMC4693185 DOI: 10.3390/metabo5040536] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/04/2015] [Accepted: 09/23/2015] [Indexed: 12/14/2022] Open
Abstract
Essentiality (ES) and Synthetic Lethality (SL) information identify combination of genes whose deletion inhibits cell growth. This information is important for both identifying drug targets for tumor and pathogenic bacteria suppression and for flagging and avoiding gene deletions that are non-viable in biotechnology. In this study, we performed a comprehensive ES and SL analysis of two important eukaryotic models (S. cerevisiae and CHO cells) using a bilevel optimization approach introduced earlier. Information gleaned from this study is used to propose specific model changes to remedy inconsistent with data model predictions. Even for the highly curated Yeast 7.11 model we identified 50 changes (metabolic and GPR) leading to the correct prediction of an additional 28% of essential genes and 36% of synthetic lethals along with a 53% reduction in the erroneous identification of essential genes. Due to the paucity of mutant growth phenotype data only 12 changes were made for the CHO 1.2 model leading to an additional correctly predicted 11 essential and eight non-essential genes. Overall, we find that CHO 1.2 was 76% less accurate than the Yeast 7.11 metabolic model in predicting essential genes. Based on this analysis, 14 (single and double deletion) maximally informative experiments are suggested to improve the CHO cell model by using information from a mouse metabolic model. This analysis demonstrates the importance of single and multiple knockout phenotypes in assessing and improving model reconstructions. The advent of techniques such as CRISPR opens the door for the global assessment of eukaryotic models.
Collapse
Affiliation(s)
- Ratul Chowdhury
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, PA 16802, USA.
| | - Anupam Chowdhury
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, PA 16802, USA.
| | - Costas D Maranas
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, PA 16802, USA.
| |
Collapse
|
86
|
Therapeutic bispecific T-cell engager antibody targeting the intracellular oncoprotein WT1. Nat Biotechnol 2015; 33:1079-86. [PMID: 26389576 DOI: 10.1038/nbt.3349] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 07/29/2015] [Indexed: 11/08/2022]
Abstract
Intracellular tumor antigens presented on the cell surface in the context of human leukocyte antigen (HLA) molecules have been targeted by T cell-based therapies, but there has been little progress in developing small-molecule drugs or antibodies directed to these antigens. Here we describe a bispecific T-cell engager (BiTE) antibody derived from a T-cell receptor (TCR)-mimic monoclonal antibody (mAb) ESK1, which binds a peptide derived from the intracellular oncoprotein WT1 presented on HLA-A*02:01. Despite the very low density of the complexes at the cell surface, ESK1-BiTE selectively activated and induced proliferation of cytolytic human T cells that killed cells from multiple leukemias and solid tumors in vitro and in mice. We also discovered that in an autologous in vitro setting, ESK1-BiTE induced a robust secondary CD8 T-cell response specific for tumor-associated antigens other than WT1. Our study provides an approach that targets tumor-specific intracellular antigens without using cell therapy and suggests that epitope spreading could contribute to the therapeutic efficacy of this BiTE.
Collapse
|
87
|
Büssow K. Stable mammalian producer cell lines for structural biology. Curr Opin Struct Biol 2015; 32:81-90. [DOI: 10.1016/j.sbi.2015.03.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/11/2015] [Accepted: 03/03/2015] [Indexed: 11/28/2022]
|
88
|
Rajendra Y, Hougland MD, Alam R, Morehead TA, Barnard GC. A high cell density transient transfection system for therapeutic protein expression based on a CHO GS-knockout cell line: process development and product quality assessment. Biotechnol Bioeng 2015; 112:977-86. [PMID: 25502369 DOI: 10.1002/bit.25514] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/05/2014] [Accepted: 12/03/2014] [Indexed: 01/15/2023]
Abstract
Transient gene expression (TGE) is a rapid method for the production of recombinant proteins in mammalian cells. While the volumetric productivity of TGE has improved significantly over the past decade, most methods involve extensive cell line engineering and plasmid vector optimization in addition to long fed batch cultures lasting up to 21 days. Our colleagues have recently reported the development of a CHO K1SV GS-KO host cell line. By creating a bi-allelic glutamine synthetase knock out of the original CHOK1SV host cell line, they were able to improve the efficiency of generating high producing stable CHO lines for drug product manufacturing. We developed a TGE method using the same CHO K1SV GS-KO host cell line without any further cell line engineering. We also refrained from performing plasmid vector engineering. Our objective was to setup a TGE process to mimic protein quality attributes obtained from stable CHO cell line. Polyethyleneimine (PEI)-mediated transfections were performed at high cell density (4 × 10(6) cells/mL) followed by immediate growth arrest at 32 °C for 7 days. Optimizing DNA and PEI concentrations proved to be important. Interestingly, found the direct transfection method (where DNA and PEI were added sequentially) to be superior to the more common indirect method (where DNA and PEI are first pre-complexed). Moreover, the addition of a single feed solution and a polar solvent (N,N dimethylacetamide) significantly increased product titers. The scalability of process from 2 mL to 2 L was demonstrated using multiple proteins and multiple expression volumes. Using this simple, short, 7-day TGE process, we were able to successfully produce 54 unique proteins in a fraction of the time that would have been required to produce the respective stable CHO cell lines. The list of 54 unique proteins includes mAbs, bispecific antibodies, and Fc-fusion proteins. Antibody titers of up to 350 mg/L were achieved with the simple 7-day process. Titers were increased to 1 g/L by extending the culture to 16 days. We also present two case studies comparing product quality of material generated by transient HEK293, transient CHO K1SV GS-KO, and stable CHO K1SV KO pool. Protein from transient CHO was more representative of stable CHO protein compared to protein produced from HEK293.
Collapse
Affiliation(s)
- Yashas Rajendra
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, 46285, Indiana
| | | | | | | | | |
Collapse
|
89
|
Overexpression of Serpinb1 in Chinese hamster ovary cells increases recombinant IgG productivity. J Biotechnol 2015; 193:91-9. [DOI: 10.1016/j.jbiotec.2014.10.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/25/2014] [Accepted: 10/31/2014] [Indexed: 11/23/2022]
|
90
|
Barnard GC, Hougland MD, Rajendra Y. High-throughput mAb expression and purification platform based on transient CHO. Biotechnol Prog 2014; 31:239-47. [DOI: 10.1002/btpr.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/10/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Gavin C. Barnard
- Biotechnology Discovery Research; Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center; Indianapolis IN 46285
| | - Maria D. Hougland
- Biotechnology Discovery Research; Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center; Indianapolis IN 46285
| | - Yashas Rajendra
- Biotechnology Discovery Research; Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center; Indianapolis IN 46285
| |
Collapse
|
91
|
Role of epigenetics in expression of recombinant proteins from mammalian cells. ACTA ACUST UNITED AC 2014. [DOI: 10.4155/pbp.14.47] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
92
|
Cheng JK, Alper HS. The genome editing toolbox: a spectrum of approaches for targeted modification. Curr Opin Biotechnol 2014; 30:87-94. [PMID: 25010940 DOI: 10.1016/j.copbio.2014.06.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 05/11/2014] [Accepted: 06/08/2014] [Indexed: 02/05/2023]
Abstract
The increase in quality, quantity, and complexity of recombinant products heavily drives the need to predictably engineer model and complex (mammalian) cell systems. However, until recently, limited tools offered the ability to precisely manipulate their genomes, thus impeding the full potential of rational cell line development processes. Targeted genome editing can combine the advances in synthetic and systems biology with current cellular hosts to further push productivity and expand the product repertoire. This review highlights recent advances in targeted genome editing techniques, discussing some of their capabilities and limitations and their potential to aid advances in pharmaceutical biotechnology.
Collapse
Affiliation(s)
- Joseph K Cheng
- McKetta Department of Chemical Engineering, The University of Texas at Austin, 200 E Dean Keeton Street, Stop C0400, Austin, TX 78712, USA
| | - Hal S Alper
- McKetta Department of Chemical Engineering, The University of Texas at Austin, 200 E Dean Keeton Street, Stop C0400, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, The University of Texas at Austin, 2500 Speedway Avenue, Austin, TX 78712, USA.
| |
Collapse
|
93
|
Steentoft C, Bennett EP, Schjoldager KTBG, Vakhrushev SY, Wandall HH, Clausen H. Precision genome editing: a small revolution for glycobiology. Glycobiology 2014; 24:663-80. [PMID: 24861053 DOI: 10.1093/glycob/cwu046] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Precise and stable gene editing in mammalian cell lines has until recently been hampered by the lack of efficient targeting methods. While different gene silencing strategies have had tremendous impact on many biological fields, they have generally not been applied with wide success in the field of glycobiology, primarily due to their low efficiencies, with resultant failure to impose substantial phenotypic consequences upon the final glycosylation products. Here, we review novel nuclease-based precision genome editing techniques enabling efficient and stable gene editing, including gene disruption, insertion, repair, modification and deletion. The nuclease-based techniques comprised of homing endonucleases, zinc finger nucleases, transcription activator-like effector nucleases, as well as the RNA-guided clustered regularly interspaced short palindromic repeat/Cas nuclease system, all function by introducing single or double-stranded breaks at a defined genomic sequence. We here compare and contrast the different techniques and summarize their current applications, highlighting cases from the field of glycobiology as well as pointing to future opportunities. The emerging potential of precision gene editing for the field is exemplified by applications to xenotransplantation; to probing O-glycoproteomes, including differential O-GalNAc glycoproteomes, to decipher the function of individual polypeptide GalNAc-transferases, as well as for engineering Chinese Hamster Ovary host cells for production of improved therapeutic biologics.
Collapse
Affiliation(s)
- Catharina Steentoft
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Eric P Bennett
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Katrine T-B G Schjoldager
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| |
Collapse
|
94
|
Complete Knockout of the Lactate Dehydrogenase A Gene is Lethal in Pyruvate Dehydrogenase Kinase 1, 2, 3 Down-Regulated CHO Cells. Mol Biotechnol 2014; 56:833-8. [DOI: 10.1007/s12033-014-9762-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
95
|
Hendel A, Kildebeck EJ, Fine EJ, Clark J, Punjya N, Sebastiano V, Bao G, Porteus MH. Quantifying genome-editing outcomes at endogenous loci with SMRT sequencing. Cell Rep 2014; 7:293-305. [PMID: 24685129 DOI: 10.1016/j.celrep.2014.02.040] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/16/2014] [Accepted: 02/26/2014] [Indexed: 12/19/2022] Open
Abstract
Targeted genome editing with engineered nucleases has transformed the ability to introduce precise sequence modifications at almost any site within the genome. A major obstacle to probing the efficiency and consequences of genome editing is that no existing method enables the frequency of different editing events to be simultaneously measured across a cell population at any endogenous genomic locus. We have developed a method for quantifying individual genome-editing outcomes at any site of interest with single-molecule real-time (SMRT) DNA sequencing. We show that this approach can be applied at various loci using multiple engineered nuclease platforms, including transcription-activator-like effector nucleases (TALENs), RNA-guided endonucleases (CRISPR/Cas9), and zinc finger nucleases (ZFNs), and in different cell lines to identify conditions and strategies in which the desired engineering outcome has occurred. This approach offers a technique for studying double-strand break repair, facilitates the evaluation of gene-editing technologies, and permits sensitive quantification of editing outcomes in almost every experimental system used.
Collapse
Affiliation(s)
- Ayal Hendel
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Eric J Kildebeck
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Eli J Fine
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Joseph Clark
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Niraj Punjya
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Vittorio Sebastiano
- Department of Obstetrics and Gynecology, Stanford University, Stanford, CA 94305, USA
| | - Gang Bao
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
96
|
Bandaranayake AD, Almo SC. Recent advances in mammalian protein production. FEBS Lett 2013; 588:253-60. [PMID: 24316512 DOI: 10.1016/j.febslet.2013.11.035] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 11/25/2013] [Accepted: 11/26/2013] [Indexed: 12/14/2022]
Abstract
Mammalian protein production platforms have had a profound impact in many areas of basic and applied research, and an increasing number of blockbuster drugs are recombinant mammalian proteins. With global sales of these drugs exceeding US$120 billion per year, both industry and academic research groups continue to develop cost effective methods for producing mammalian proteins to support pre-clinical and clinical evaluations of potential therapeutics. While a wide range of platforms have been successfully exploited for laboratory use, the bulk of recent biologics have been produced in mammalian cell lines due to the requirement for post translational modification and the biosynthetic complexity of the target proteins. In this review we highlight the range of mammalian expression platforms available for recombinant protein production, as well as advances in technologies for the rapid and efficient selection of highly productive clones.
Collapse
Affiliation(s)
- Ashok D Bandaranayake
- Departments of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| | - Steven C Almo
- Departments of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States; Physiology and Biophysics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| |
Collapse
|
97
|
Development of a highly-efficient CHO cell line generation system with engineered SV40E promoter. J Biotechnol 2013; 168:652-8. [DOI: 10.1016/j.jbiotec.2013.08.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 08/05/2013] [Accepted: 08/16/2013] [Indexed: 11/17/2022]
|
98
|
The use of glutamine synthetase as a selection marker: recent advances in Chinese hamster ovary cell line generation processes. ACTA ACUST UNITED AC 2013. [DOI: 10.4155/pbp.13.56] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
99
|
Noh SM, Sathyamurthy M, Lee GM. Development of recombinant Chinese hamster ovary cell lines for therapeutic protein production. Curr Opin Chem Eng 2013. [DOI: 10.1016/j.coche.2013.08.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
100
|
Richter S, Morrison S, Connor T, Su J, Print CG, Ronimus RS, McGee SL, Wilson WR. Zinc finger nuclease mediated knockout of ADP-dependent glucokinase in cancer cell lines: effects on cell survival and mitochondrial oxidative metabolism. PLoS One 2013; 8:e65267. [PMID: 23799003 PMCID: PMC3683018 DOI: 10.1371/journal.pone.0065267] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 04/23/2013] [Indexed: 01/22/2023] Open
Abstract
Zinc finger nucleases (ZFN) are powerful tools for editing genes in cells. Here we use ZFNs to interrogate the biological function of ADPGK, which encodes an ADP-dependent glucokinase (ADPGK), in human tumour cell lines. The hypothesis we tested is that ADPGK utilises ADP to phosphorylate glucose under conditions where ATP becomes limiting, such as hypoxia. We characterised two ZFN knockout clones in each of two lines (H460 and HCT116). All four clones had frameshift mutations in all alleles at the target site in exon 1 of ADPGK, and were ADPGK-null by immunoblotting. ADPGK knockout had little or no effect on cell proliferation, but compromised the ability of H460 cells to survive siRNA silencing of hexokinase-2 under oxic conditions, with clonogenic survival falling from 21±3% for the parental line to 6.4±0.8% (p = 0.002) and 4.3±0.8% (p = 0.001) for the two knockouts. A similar increased sensitivity to clonogenic cell killing was observed under anoxia. No such changes were found when ADPGK was knocked out in HCT116 cells, for which the parental line was less sensitive than H460 to anoxia and to hexokinase-2 silencing. While knockout of ADPGK in HCT116 cells caused few changes in global gene expression, knockout of ADPGK in H460 cells caused notable up-regulation of mRNAs encoding cell adhesion proteins. Surprisingly, we could discern no consistent effect on glycolysis as measured by glucose consumption or lactate formation under anoxia, or extracellular acidification rate (Seahorse XF analyser) under oxic conditions in a variety of media. However, oxygen consumption rates were generally lower in the ADPGK knockouts, in some cases markedly so. Collectively, the results demonstrate that ADPGK can contribute to tumour cell survival under conditions of high glycolytic dependence, but the phenotype resulting from knockout of ADPGK is cell line dependent and appears to be unrelated to priming of glycolysis in these lines.
Collapse
Affiliation(s)
- Susan Richter
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Shona Morrison
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Tim Connor
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Jiechuang Su
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Cristin G. Print
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- The New Zealand Bioinformatics Institute, The University of Auckland, Auckland, New Zealand
| | | | - Sean L. McGee
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - William R. Wilson
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- * E-mail:
| |
Collapse
|