1
|
Boukari I, Rourou S, Bouzazi D, Essafi-Benkhadir K, Kallel H. Strategies for improving expression of recombinant human chorionic gonadotropin in Chinese Hamster Ovary (CHO) cells. Protein Expr Purif 2025; 225:106596. [PMID: 39218246 DOI: 10.1016/j.pep.2024.106596] [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: 06/17/2024] [Revised: 08/14/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Optimizations of the gene expression cassette combined with the selection of an appropriate signal peptide are important factors that must be considered to enhance heterologous protein expression in Chinese Hamster Ovary (CHO) cells. In this study, we investigated the effectiveness of different signal peptides on the production of recombinant human chorionic gonadotropin (r-hCG) in CHO-K1 cells. Four optimized expression constructs containing four promising signal peptides were stably transfected into CHO-K1 cells. The generated CHO-K1 stable pool was then evaluated for r-hCG protein production. Interestingly, human serum albumin and human interleukin-2 signal peptides exhibited relatively greater extracellular secretion of the r-hCG with an average yield of (16.59 ± 0.02 μg/ml) and (14.80 ± 0.13 μg/ml) respectively compared to the native and murine IgGκ light chain signal peptides. The stably transfected CHO pool was further used as the cell substrate to develop an optimized upstream process followed by a downstream phase of the r-hCG. Finally, the biological activity of the purified r-hCG was assessed using in vitro bioassays. The combined data highlight that the choice of signal peptide can be imperative to ensure an optimal secretion of a recombinant protein in CHO cells. In addition, the stable pool technology was a viable approach for the production of biologically active r-hCG at a research scale with acceptable bioprocess performances and consistent product quality.
Collapse
Affiliation(s)
- Iheb Boukari
- Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, LR16IPT01, Biotechnology Development Group, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia; Faculty of Sciences of Bizerte, Carthage University, 7021, Bizerte, Tunisia.
| | - Samia Rourou
- Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, LR16IPT01, Biotechnology Development Group, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia
| | - Dorsaf Bouzazi
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia
| | - Khadija Essafi-Benkhadir
- Laboratory of Molecular Epidemiology and Experimental Pathology, LR16IPT04, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia
| | - Héla Kallel
- Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, LR16IPT01, Biotechnology Development Group, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia; Univercells SA, Belgium
| |
Collapse
|
2
|
Gonzalez-Rivera JC, Galvan A, Ryder T, Milman M, Agarwal K, Kandari L, Khetan A. A high-titer scalable Chinese hamster ovary transient expression platform for production of biotherapeutics. Biotechnol Bioeng 2024; 121:3454-3470. [PMID: 39101569 DOI: 10.1002/bit.28817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 07/08/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024]
Abstract
Transient gene expression (TGE) in Chinese hamster ovary (CHO) cells offers a route to accelerate biologics development by delivering material weeks to months earlier than what is possible with conventional cell line development. However, low productivity, inconsistent product quality profiles, and scalability challenges have prevented its broader adoption. In this study, we develop a scalable CHO-based TGE system achieving 1.9 g/L of monoclonal antibody in an unmodified host. We integrated continuous flow-electroporation and alternate tangential flow (ATF) perfusion to enable an end-to-end closed system from N-1 perfusion to fed-batch 50-L bioreactor production. Optimization of both the ATF operation for three-in-one application-cell growth, buffer exchange, and cell mass concentration-and the flow-electroporation process, led to a platform for producing biotherapeutics using transiently transfected cells. We demonstrate scalability up to 50-L bioreactor, maintaining a titer over 1 g/L. We also show comparable quality between both transiently and stably produced material, and consistency across batches. The results confirm that purity, charge variants and N-glycan profiles are similar. Our study demonstrates the potential of CHO-based TGE platforms to accelerate biologics process development timelines and contributes evidence supporting its feasibility for manufacturing early clinical material, aiming to strengthen endorsement for TGE's wider implementation.
Collapse
Affiliation(s)
| | - Alberto Galvan
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Todd Ryder
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Monica Milman
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Kitty Agarwal
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Lakshmi Kandari
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Anurag Khetan
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| |
Collapse
|
3
|
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
|
4
|
Peltret M, Vetsch P, Farvaque E, Mette R, Tsachaki M, Duarte L, Duret A, Vaxelaire E, Frank J, Moritz B, Aillerie C, Giovannini R, Bertschinger M. Development of a 10 g/L process for a difficult-to-express multispecific antibody format using a holistic process development approach. J Biotechnol 2024; 389:30-42. [PMID: 38685416 DOI: 10.1016/j.jbiotec.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
Ichnos has developed a multi-specific antibody platform based on the BEAT® (Bispecific engagement by antibodies based on the T-cell receptor) interface. The increased complexity of the bi- and multi-specific formats generated with this platform makes these molecules difficult-to-express proteins compared to standard monoclonal antibodies (mAbs). This report describes how expression limitations of a bi-specific bi-paratopic BEAT antibody were improved in a holistic approach. An initial investigation allowed identification of a misbalance in the subunits composing the BEAT antibody as the potential root cause. This misbalance was then addressed by a signal peptide optimization, and the overall expression level was increased by the combination of two vector design elements on a single gene vector. Further improvements were made in the selection of cell populations and an upstream (USP) platform process was applied in combination with a cell culture temperature shift. This allowed titer levels of up to 6 g/L to be reached with these difficult-to-express proteins. Furthermore, a high-density seeding process was developed that allowed titers of around 11 g/L for the BEAT antibody, increasing the initial titer by a factor of 10. The approach was successfully applied to a tri-specific antibody with titer levels reaching 10 g/L. In summary, a platform process for difficult-to-express proteins was developed using molecular biology tools, cell line development, upstream process optimization and process intensification.
Collapse
Affiliation(s)
- Mégane Peltret
- Drug Substance Development, Ichnos Sciences, Switzerland
| | - Patrick Vetsch
- Drug Substance Development, Ichnos Sciences, Switzerland
| | | | - Romain Mette
- Drug Substance Development, Ichnos Sciences, Switzerland
| | - Maria Tsachaki
- Drug Substance Development, Ichnos Sciences, Switzerland
| | - Lionel Duarte
- Drug Substance Development, Ichnos Sciences, Switzerland
| | - Anaïs Duret
- Drug Substance Development, Ichnos Sciences, Switzerland
| | | | - Jana Frank
- Drug Substance Development, Ichnos Sciences, Switzerland
| | | | | | | | | |
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
|
Gaa R, Kumari K, Mayer HM, Yanakieva D, Tsai SP, Joshi S, Guenther R, Doerner A. An integrated mammalian library approach for optimization and enhanced microfluidics-assisted antibody hit discovery. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:74-82. [PMID: 36762883 DOI: 10.1080/21691401.2023.2173219] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Recent years have seen the development of a variety of mammalian library approaches for display and secretion mode. Advantages include library approaches for engineering, preservation of precious immune repertoires and their repeated interrogation, as well as screening in final therapeutic format and host. Mammalian display approaches for antibody optimization exploit these advantages, necessitating the generation of large libraries but in turn enabling early screening for both manufacturability and target specificity. For suitable libraries, high antibody integration rates and resulting monoclonality need to be balanced - we present a solution for sufficient transmutability and acceptable monoclonality by applying an optimized ratio of coding to non-coding lentivirus. The recent advent of microfluidic-assisted hit discovery represents a perfect match to mammalian libraries in secretion mode, as the lower throughput fits well with the facile generation of libraries comprising a few million functional clones. In the presented work, Chinese Hamster Ovary cells were engineered to both express the target of interest and secrete antibodies in relevant formats, and specific clones were strongly enriched by high throughput screening for autocrine cellular binding. The powerful combination of mammalian secretion libraries and microfluidics-assisted hit discovery could reduce attrition rates and increase the probability to identify the best possible therapeutic antibody hits faster.
Collapse
Affiliation(s)
- Ramona Gaa
- Protein Engineering and Antibody Technologies, Merck KGaA, Darmstadt, Germany
| | - Kavita Kumari
- Discovery Biology, Syngene International, Phase-IV, Bangalore, India
| | - Hannah Melina Mayer
- Protein Engineering and Antibody Technologies, Merck KGaA, Darmstadt, Germany
| | - Desislava Yanakieva
- Protein Engineering and Antibody Technologies, Merck KGaA, Darmstadt, Germany
| | - Shang-Pu Tsai
- Protein Engineering and Antibody Technologies, EMD Serono, Billerica, MA, USA
| | - Saurabh Joshi
- Discovery Biology, Syngene International, Phase-IV, Bangalore, India
| | - Ralf Guenther
- Protein Engineering and Antibody Technologies, Merck KGaA, Darmstadt, Germany
| | - Achim Doerner
- Protein Engineering and Antibody Technologies, Merck KGaA, Darmstadt, Germany
| |
Collapse
|
7
|
Sun H, Wang S, Lu M, Tinberg CE, Alba BM. Protein production from HEK293 cell line-derived stable pools with high protein quality and quantity to support discovery research. PLoS One 2023; 18:e0285971. [PMID: 37267316 DOI: 10.1371/journal.pone.0285971] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/07/2023] [Indexed: 06/04/2023] Open
Abstract
Antibody-based therapeutics and recombinant protein reagents are often produced in mammalian expression systems, which provide human-like post-translational modifications. Among the available mammalian cell lines used for recombinant protein expression, Chinese hamster ovary (CHO)-derived suspension cells are generally utilized because they are easy to culture and tend to produce proteins in high yield. However, some proteins purified from CHO cell overexpression suffer from clipping and display undesired non-human post translational modifications (PTMs). In addition, CHO cell lines are often not suitable for producing proteins with many glycosylation motifs for structural biology studies, as N-linked glycosylation of proteins poses challenges for structure determination by X-ray crystallography. Hence, alternative and complementary cell lines are required to address these issues. Here, we present a robust method for expressing proteins in human embryonic kidney 293 (HEK293)-derived stable pools, leading to recombinant protein products with much less clipped species compared to those expressed in CHO cells and with higher yield compared to those expressed in transiently-transfected HEK293 cells. Importantly, the stable pool generation protocol is also applicable to HEK293S GnTI- (N-acetylglucosaminyltransferase I-negative) and Expi293F GnTI- suspension cells, facilitating production of high yields of proteins with less complex glycans for use in structural biology projects. Compared to HEK293S GnTI- stable pools, Expi293F GnTI- stable pools consistently produce proteins with similar or higher expression levels. HEK293-derived stable pools can lead to a significant cost reduction and greatly promote the production of high-quality proteins for diverse research projects.
Collapse
Affiliation(s)
- Hong Sun
- Biologic Therapeutic Discovery, Amgen Research, South San Francisco, California, United States of America
| | - Songyu Wang
- Biologic Therapeutic Discovery, Amgen Research, South San Francisco, California, United States of America
| | - Mei Lu
- Biologic Therapeutic Discovery, Amgen Research, South San Francisco, California, United States of America
| | - Christine E Tinberg
- Biologic Therapeutic Discovery, Amgen Research, South San Francisco, California, United States of America
| | - Benjamin M Alba
- Biologic Therapeutic Discovery, Amgen Research, South San Francisco, California, United States of America
| |
Collapse
|
8
|
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
|
9
|
Oliviero C, Hinz SC, Bogen JP, Kornmann H, Hock B, Kolmar H, Hagens G. Generation of a Host Cell line containing a MAR-rich landing pad for site-specific integration and expression of transgenes. Biotechnol Prog 2022; 38:e3254. [PMID: 35396920 PMCID: PMC9539524 DOI: 10.1002/btpr.3254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/22/2022] [Accepted: 03/31/2022] [Indexed: 11/10/2022]
Abstract
In recent years, targeted gene integration (TI) has been introduced as a strategy for the generation of recombinant mammalian cell lines for the production of biotherapeutics. Besides reducing the immense heterogeneity within a pool of recombinant transfectants, TI also aims at shortening the duration of the current cell line development process. Here we describe the generation of a host cell line carrying Matrix‐Attachment Region (MAR)‐rich landing pads (LPs), which allow for the simultaneous and site‐specific integration of multiple genes of interest (GOIs). We show that several copies of each chicken lysozyme 5'MAR‐based LP containing either BxB1 wild type or mutated recombination sites, integrated at one random chromosomal locus of the host cell genome. We further demonstrate that these LP‐containing host cell lines can be used for the site‐specific integration of several GOIs and thus, generation of transgene‐expressing stable recombinant clones. Transgene expression was shown by site‐specific integration of heavy and light chain genes coding for a monospecific antibody (msAb) as well as for a bi‐specific antibody (bsAb). The genetic stability of the herein described LP‐based recombinant clones expressing msAb or bsAb was demonstrated by cultivating the recombinant clones and measuring antibody titers over 85 generations. We conclude that the host cell containing multiple copies of MAR‐rich landing pads can be successfully used for stable expression of one or several GOIs.
Collapse
Affiliation(s)
- Claudia Oliviero
- Institute of Life Technology, Haute Ecole d'Ingénierie HES-SO Valais Wallis, Rue de l'Industrie 19, CH-1950 Sion, Switzerland
| | - Steffen C Hinz
- Institute of Life Technology, Haute Ecole d'Ingénierie HES-SO Valais Wallis, Rue de l'Industrie 19, CH-1950 Sion, Switzerland
| | - Jan P Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, D-64287, Darmstadt, Germany
| | - Henri Kornmann
- Ferring Biologics Innovation Center, Route de la Corniche 8, CH-1066, Epalinges, Switzerland
| | - Björn Hock
- Ferring Biologics Innovation Center, Route de la Corniche 8, CH-1066, Epalinges, Switzerland.,SwissThera SA, Route de la Corniche 4, CH-1066, Epalinges, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, D-64287, Darmstadt, Germany
| | - Gerrit Hagens
- Institute of Life Technology, Haute Ecole d'Ingénierie HES-SO Valais Wallis, Rue de l'Industrie 19, CH-1950 Sion, Switzerland
| |
Collapse
|
10
|
Tejwani V, Chaudhari M, Rai T, Sharfstein ST. High-throughput and automation advances for accelerating single-cell cloning, monoclonality and early phase clone screening steps in mammalian cell line development for biologics production. Biotechnol Prog 2021; 37:e3208. [PMID: 34478248 DOI: 10.1002/btpr.3208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
Mammalian cell line development is a multistep process wherein timelines for developing clonal cells to be used as manufacturing cell lines for biologics production can commonly extend to 9 months when no automation or modern molecular technologies are involved in the workflow. Steps in the cell line development workflow involving single-cell cloning, monoclonality assurance, productivity and stability screening are labor, time and resource intensive when performed manually. Introduction of automation and miniaturization in these steps has reduced the required manual labor, shortened timelines from months to weeks, and decreased the resources needed to develop manufacturing cell lines. This review summarizes the advances, benefits, comparisons and shortcomings of different automation platforms available in the market for rapid isolation of desired clonal cell lines for biologics production.
Collapse
Affiliation(s)
- Vijay Tejwani
- Biotechnology R&D, Clone Development Team, Lupin Limited, Pune, India
| | - Minal Chaudhari
- Biotechnology R&D, Clone Development Team, Lupin Limited, Pune, India
| | - Toyaj Rai
- Biotechnology R&D, Clone Development Team, Lupin Limited, Pune, India
| | - Susan T Sharfstein
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York, USA
| |
Collapse
|
11
|
Napoleone A, Laurén I, Linkgreim T, Dahllund L, Persson H, Andersson O, Olsson A, Hultqvist G, Frank P, Hall M, Morrison A, Andersson A, Lord M, Mangsbo S. Fed-batch production assessment of a tetravalent bispecific antibody: A case study on piggyBac stably transfected HEK293 cells. N Biotechnol 2021; 65:9-19. [PMID: 34273575 DOI: 10.1016/j.nbt.2021.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/07/2021] [Accepted: 07/11/2021] [Indexed: 12/24/2022]
Abstract
The transition from preclinical biological drug development into clinical trials requires an efficient upscaling process. In this context, bispecific antibody drugs are particularly challenging due to their propensity to form aggregates and generally produce low titers. Here, the upscaling process for a tetravalent bispecific antibody expressed by a piggyBac transposon-mediated stable HEK293 cell pool has been evaluated. The project was performed as a case study at Testa Center, a non-GMP facility for scale-up testing of biologics in Sweden, and encompassed media adaptation strategies, fed-batch optimization and a novel antibody purification technology. The cell pool was adapted to different culture media for evaluation in terms of cell viability and titers compared to its original Expi293 Expression Medium. These parameters were assessed in both sequential stepwise adaption and direct media exchanges. By this, a more affordable medium was identified that did not require stepwise adaptation and with similar titers and viability as in the Expi293 Expression Medium. Fed-batch optimizations resulted in culture densities reaching up to 20 × 106 viable cells/mL with over 90 % viability 12 days post-inoculum, and antibody titers three times higher than corresponding batch cultures. By implementing a novel high-speed protein A fiber technology (Fibro PrismA) with a capture residence time of only 7.5 s, 8 L of supernatant could be purified in 4.5 h without compromising the purity, structural integrity and function of the bispecific antibody. Results from this study related to medium adaptation and design of fed-batch protocols will be highly beneficial during the forthcoming scale-up of this therapeutic antibody.
Collapse
Affiliation(s)
- Antonino Napoleone
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ida Laurén
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Triinu Linkgreim
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Leif Dahllund
- Science for Life Laboratory, Drug Discovery and Development & School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Helena Persson
- Science for Life Laboratory, Drug Discovery and Development & School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Oskar Andersson
- Science for Life Laboratory, Drug Discovery and Development & School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anders Olsson
- Science for Life Laboratory, Drug Discovery and Development & School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Greta Hultqvist
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Peter Frank
- Wicket AB, Väderkvarnsgatan 30, 75329 Uppsala, Sweden
| | - Martin Hall
- Cytiva AB, Björkgatan 30, 751 84 Uppsala, Sweden
| | - Annika Morrison
- Cytiva Testa Center AB, Björkgatan 30, 751 84 Uppsala, Sweden
| | | | - Martin Lord
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
12
|
Jones BE, Brown-Augsburger PL, Corbett KS, Westendorf K, Davies J, Cujec TP, Wiethoff CM, Blackbourne JL, Heinz BA, Foster D, Higgs RE, Balasubramaniam D, Wang L, Zhang Y, Yang ES, Bidshahri R, Kraft L, Hwang Y, Žentelis S, Jepson KR, Goya R, Smith MA, Collins DW, Hinshaw SJ, Tycho SA, Pellacani D, Xiang P, Muthuraman K, Sobhanifar S, Piper MH, Triana FJ, Hendle J, Pustilnik A, Adams AC, Berens SJ, Baric RS, Martinez DR, Cross RW, Geisbert TW, Borisevich V, Abiona O, Belli HM, de Vries M, Mohamed A, Dittmann M, Samanovic MI, Mulligan MJ, Goldsmith JA, Hsieh CL, Johnson NV, Wrapp D, McLellan JS, Barnhart BC, Graham BS, Mascola JR, Hansen CL, Falconer E. The neutralizing antibody, LY-CoV555, protects against SARS-CoV-2 infection in nonhuman primates. Sci Transl Med 2021; 13:eabf1906. [PMID: 33820835 PMCID: PMC8284311 DOI: 10.1126/scitranslmed.abf1906] [Citation(s) in RCA: 313] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/19/2021] [Accepted: 03/31/2021] [Indexed: 12/15/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) poses a public health threat for which preventive and therapeutic agents are urgently needed. Neutralizing antibodies are a key class of therapeutics that may bridge widespread vaccination campaigns and offer a treatment solution in populations less responsive to vaccination. Here, we report that high-throughput microfluidic screening of antigen-specific B cells led to the identification of LY-CoV555 (also known as bamlanivimab), a potent anti-spike neutralizing antibody from a hospitalized, convalescent patient with coronavirus disease 2019 (COVID-19). Biochemical, structural, and functional characterization of LY-CoV555 revealed high-affinity binding to the receptor-binding domain, angiotensin-converting enzyme 2 binding inhibition, and potent neutralizing activity. A pharmacokinetic study of LY-CoV555 conducted in cynomolgus monkeys demonstrated a mean half-life of 13 days and a clearance of 0.22 ml hour-1 kg-1, consistent with a typical human therapeutic antibody. In a rhesus macaque challenge model, prophylactic doses as low as 2.5 mg/kg reduced viral replication in the upper and lower respiratory tract in samples collected through study day 6 after viral inoculation. This antibody has entered clinical testing and is being evaluated across a spectrum of COVID-19 indications, including prevention and treatment.
Collapse
Affiliation(s)
- Bryan E Jones
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA.
| | | | - Kizzmekia S Corbett
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Julian Davies
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Thomas P Cujec
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | | | | | | | - Denisa Foster
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | | | | | - Lingshu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yi Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eun Sung Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Lucas Kraft
- AbCellera Biologics Inc., Vancouver, BC V5Y0A1, Canada
| | - Yuri Hwang
- AbCellera Biologics Inc., Vancouver, BC V5Y0A1, Canada
| | | | | | - Rodrigo Goya
- AbCellera Biologics Inc., Vancouver, BC V5Y0A1, Canada
| | - Maia A Smith
- AbCellera Biologics Inc., Vancouver, BC V5Y0A1, Canada
| | | | | | - Sean A Tycho
- AbCellera Biologics Inc., Vancouver, BC V5Y0A1, Canada
| | | | - Ping Xiang
- AbCellera Biologics Inc., Vancouver, BC V5Y0A1, Canada
| | | | | | - Marissa H Piper
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Franz J Triana
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Jorg Hendle
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | - Anna Pustilnik
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA
| | | | | | - Ralph S Baric
- University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David R Martinez
- University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Robert W Cross
- Galveston National Laboratory and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thomas W Geisbert
- Galveston National Laboratory and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Olubukola Abiona
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hayley M Belli
- Department of Population Health, Division of Biostatistics, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Maren de Vries
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Adil Mohamed
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Meike Dittmann
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Marie I Samanovic
- NYU Langone Vaccine Center, Department of Medicine, Division of Infectious Diseases and Immunology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Mark J Mulligan
- NYU Langone Vaccine Center, Department of Medicine, Division of Infectious Diseases and Immunology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jory A Goldsmith
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Ching-Lin Hsieh
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Nicole V Johnson
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Daniel Wrapp
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | | | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carl L Hansen
- AbCellera Biologics Inc., Vancouver, BC V5Y0A1, Canada
| | | |
Collapse
|
13
|
Tschorn N, Berg K, Stitz J. Transposon vector-mediated stable gene transfer for the accelerated establishment of recombinant mammalian cell pools allowing for high-yield production of biologics. Biotechnol Lett 2020; 42:1103-1112. [PMID: 32323079 PMCID: PMC7275939 DOI: 10.1007/s10529-020-02889-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/13/2020] [Indexed: 12/11/2022]
Abstract
Stable recombinant mammalian cells are of growing importance in pharmaceutical biotechnology production scenarios for biologics such as monoclonal antibodies, growth and blood factors, cytokines and subunit vaccines. However, the establishment of recombinant producer cells using classical stable transfection of plasmid DNA is hampered by low stable gene transfer efficiencies. Consequently, subsequent selection of transgenic cells and the screening of clonal cell populations are time- and thus cost-intensive. To overcome these limitations, expression cassettes were embedded into transposon-derived donor vectors. Upon the co-transfection with transposase-encoding constructs, elevated vector copy numbers stably integrated into the genomes of the host cells are readily achieved facilitating under stringent selection pressure the establishment of cell pools characterized by sustained and high-yield recombinant protein production. Here, we discuss some aspects of transposon vector technologies, which render these vectors promising candidates for their further utilization in the production of biologics.
Collapse
Affiliation(s)
- Natalie Tschorn
- Research Group Pharmaceutical Biotechnology, TH Köln - University of Applied Sciences, Chempark Leverkusen E28, Kaiser-Wilhelm-Allee, 51368, Leverkusen, Germany.,Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Karen Berg
- Research Group Pharmaceutical Biotechnology, TH Köln - University of Applied Sciences, Chempark Leverkusen E28, Kaiser-Wilhelm-Allee, 51368, Leverkusen, Germany.,Research Group Translational Hepatology and Stem Cell Biology, Cluster of Excellence REBIRTH, Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jörn Stitz
- Research Group Pharmaceutical Biotechnology, TH Köln - University of Applied Sciences, Chempark Leverkusen E28, Kaiser-Wilhelm-Allee, 51368, Leverkusen, Germany.
| |
Collapse
|
14
|
Schweickert PG, Cheng Z. Application of Genetic Engineering in Biotherapeutics Development. J Pharm Innov 2019. [DOI: 10.1007/s12247-019-09411-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
15
|
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
|
16
|
Puente-Massaguer E, Badiella L, Gutiérrez-Granados S, Cervera L, Gòdia F. A statistical approach to improve compound screening in cell culture media. Eng Life Sci 2019; 19:315-327. [PMID: 32625011 DOI: 10.1002/elsc.201800168] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/10/2019] [Accepted: 02/13/2019] [Indexed: 12/12/2022] Open
Abstract
The Chinese hamster ovary (CHO) cell line is widely used for the production of recombinant proteins due to its high growing capacity and productivity, as well as other cell lines derived later than CHO. Adapting cell culture media for each specific cell line is a key to exploit these features for cost effective and fast product generation. Media supplementation is generally addressed by means of one-factor-at-a-time or classical design of experiments approaches but these techniques may not be efficient enough in preliminary screening phases. In this study, a novel strategy consisting in folding over the Plackett-Burman design was used to increase cell growth and trastuzumab production of different CHO cell lines through supplementation with nonanimal recombinant compounds. Synergies between compounds could be detected with a reduced number of experiments by using this methodology in comparison to more conventional fractional factorial designs. In the particular case reported here, the sequential use of this modified Plackett-Burman in combination with a Box-Behnken design led to a 1.5-fold increase in cell growth (10 × 106 cells/mL) and a two-fold in trastuzumab titer (122 mg/L) in suspension batch culture.
Collapse
Affiliation(s)
- Eduard Puente-Massaguer
- Departament d'Enginyeria Química Biològica i Ambiental Campus Bellaterra Cerdanyola del Vallès Universitat Autònoma de Barcelona Barcelona Spain
| | - Llorenç Badiella
- Servei d'Estadística Aplicada Campus Bellaterra Cerdanyola del Vallès Universitat Autònoma de Barcelona Barcelona Spain
| | - Sonia Gutiérrez-Granados
- Departament d'Enginyeria Química Biològica i Ambiental Campus Bellaterra Cerdanyola del Vallès Universitat Autònoma de Barcelona Barcelona Spain
| | - Laura Cervera
- Departament d'Enginyeria Química Biològica i Ambiental Campus Bellaterra Cerdanyola del Vallès Universitat Autònoma de Barcelona Barcelona Spain
| | - Francesc Gòdia
- Departament d'Enginyeria Química Biològica i Ambiental Campus Bellaterra Cerdanyola del Vallès Universitat Autònoma de Barcelona Barcelona Spain
| |
Collapse
|
17
|
Yang O, Prabhu S, Ierapetritou M. Comparison between Batch and Continuous Monoclonal Antibody Production and Economic Analysis. Ind Eng Chem Res 2019. [DOI: 10.1021/acs.iecr.8b04717] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Ou Yang
- Department of Chemical and Biochemical Engineering, Rutgers—The State University of New Jersey, 98 Brett Road, Piscataway, New Jersey 08854-8058, United States
| | - Siddharth Prabhu
- Department of Chemical and Biochemical Engineering, Rutgers—The State University of New Jersey, 98 Brett Road, Piscataway, New Jersey 08854-8058, United States
| | - Marianthi Ierapetritou
- Department of Chemical and Biochemical Engineering, Rutgers—The State University of New Jersey, 98 Brett Road, Piscataway, New Jersey 08854-8058, United States
| |
Collapse
|
18
|
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]
|
19
|
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
|
20
|
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
|
21
|
Abstract
CHO cell pools with desirable characteristics of high titer and consistent product quality are useful for rapid production of recombinant proteins. Here we describe the generation of CHO cell pools using the piggyBac transposon system for mediating gene integration. The method describes the co-transfection of cells with the donor plasmid (coding for the gene of interest) and the helper plasmid (coding for the transposase) using polyethyleneimine (PEI). This is followed by a genetic selection for the generation of a cell pool. The resulting cell pool can be used to start a batch or fed-batch culture. Alternatively it can be used for generation of clonal cell lines or generation of cell banks for future use.
Collapse
Affiliation(s)
- Sowmya Balasubramanian
- Laboratory of Cellular Biotechnology (LBTC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland. .,ATUM, Newark, CA, USA.
| |
Collapse
|
22
|
Rajendra Y, Balasubramanian S, McCracken NA, Norris DL, Lian Z, Schmitt MG, Frye CC, Barnard GC. Evaluation of piggyBac-mediated CHO pools to enable material generation to support GLP toxicology studies. Biotechnol Prog 2017; 33:1436-1448. [DOI: 10.1002/btpr.2495] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/13/2017] [Indexed: 11/12/2022]
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
| | - 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
| | - Zhirui Lian
- Bioprocess Research and Development, Eli Lilly and Company, LTC-North; 1200 Kentucky Avenue, Indianapolis IN 46221
| | - Matthew G. Schmitt
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company; Lilly Corporate Center, Indianapolis IN 46225
| | - 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
|
23
|
Ahmadi S, Davami F, Davoudi N, Nematpour F, Ahmadi M, Ebadat S, Azadmanesh K, Barkhordari F, Mahboudi F. Monoclonal antibodies expression improvement in CHO cells by PiggyBac transposition regarding vectors ratios and design. PLoS One 2017; 12:e0179902. [PMID: 28662065 PMCID: PMC5491063 DOI: 10.1371/journal.pone.0179902] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/06/2017] [Indexed: 12/22/2022] Open
Abstract
Establishing stable Chinese Hamster Ovary (CHO) cells producing monoclonal antibodies (mAbs) usually pass through the random integration of vectors to the cell genome, which is sensitive to gene silencing. One approach to overcome this issue is to target a highly transcribed region in the genome. Transposons are useful devices to target active parts of genomes, and PiggyBac (PB) transposon can be considered as a good option. In the present study, three PB transposon donor vectors containing both heavy and light chains were constructed, one contained independent expression cassettes while the others utilized either an Internal Ribosome Entry Site (IRES) or 2A element to express mAb. Conventional cell pools were created by transferring donor vectors into the CHO cells, whereas transposon-based cells were generated by transfecting the cells with donor vectors with a companion of a transposase-encoding helper vector, with 1:2.5 helper/donor vectors ratio. To evaluate the influence of helper/donor vectors ratio on expression, the second transposon-based cell pools were generated with 1:5 helper/donor ratio. Expression levels in the transposon-based cells were two to five -folds more than those created by conventional method except for the IRES-mediated ones, in which the observed difference increased more than 100-fold. The results were dependent on both donor vector design and vectors ratios.
Collapse
Affiliation(s)
- Samira Ahmadi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Davami
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Noushin Davoudi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Nematpour
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Maryam Ahmadi
- Medical Biotechnology Department, Semnan University of Medical Sciences, Semnan, Iran
| | - Saeedeh Ebadat
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Fereidoun Mahboudi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- * E-mail: ,
| |
Collapse
|
24
|
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
|
25
|
Lalonde ME, Durocher Y. Therapeutic glycoprotein production in mammalian cells. J Biotechnol 2017; 251:128-140. [DOI: 10.1016/j.jbiotec.2017.04.028] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 04/12/2017] [Accepted: 04/23/2017] [Indexed: 12/12/2022]
|
26
|
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
|
27
|
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: 20] [Impact Index Per Article: 2.5] [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
|
28
|
Takagi Y, Kikuchi T, Wada R, Omasa T. The enhancement of antibody concentration and achievement of high cell density CHO cell cultivation by adding nucleoside. Cytotechnology 2017; 69:511-521. [PMID: 28251404 DOI: 10.1007/s10616-017-0066-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/05/2017] [Indexed: 01/05/2023] Open
Abstract
Recently, with the dramatic increase in demand for therapeutic antibodies, Chinese hamster ovary (CHO) cell culture systems have made significant progress in recombinant antibody production. Over the past two decades, recombinant antibody productivity has been improved by more than 100-fold. Medium optimization has been identified as an important key approach for increasing product concentrations. In this study, we evaluated the effects of deoxyuridine addition to fed-batch cultures of antibody-expressing CHO cell lines. Furthermore, we investigated the effects of combined addition of deoxyuridine, thymidine, and deoxycytidine. Our results suggest that addition of these pyrimidine nucleosides can increase CHO cell growth, with no significant change in the specific production rate. As a result of the increased cell growth, the antibody concentration was elevated and we were able to achieve more than 9 g/L during 16 days of culture. Similar effects of nucleoside addition were observed in fed-batch cultures of a Fab fragment-expressing CHO cell line, and the final Fab fragment concentration was more than 4 g/L. This nucleoside addition strategy could be a powerful platform for efficient antibody production.
Collapse
Affiliation(s)
- Yasuhiro Takagi
- Institute of Bioscience and Bioindustry, Tokushima University, 2-1 Minamijosanjima-cho, Tokushima, 770-8506, Japan
- Biotechnology Laboratories, Astellas Pharma Inc., 5-2-3 Tokodai, Tsukuba, Ibaraki, 300-2698, Japan
| | - Takuya Kikuchi
- Biotechnology Laboratories, Astellas Pharma Inc., 5-2-3 Tokodai, Tsukuba, Ibaraki, 300-2698, Japan
| | - Ryuta Wada
- Biotechnology Laboratories, Astellas Pharma Inc., 5-2-3 Tokodai, Tsukuba, Ibaraki, 300-2698, Japan
| | - Takeshi Omasa
- Institute of Bioscience and Bioindustry, Tokushima University, 2-1 Minamijosanjima-cho, Tokushima, 770-8506, Japan.
- Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
29
|
Xu S, Gavin J, Jiang R, Chen H. Bioreactor productivity and media cost comparison for different intensified cell culture processes. Biotechnol Prog 2017; 33:867-878. [DOI: 10.1002/btpr.2415] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/06/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Sen Xu
- Process Development and Engineering; Biologics and Vaccines, Merck & Co., Inc; 2000 Galloping Hill Road Kenilworth NJ 07033
| | - John Gavin
- Process Development and Engineering; Biologics and Vaccines, Merck & Co., Inc; 2000 Galloping Hill Road Kenilworth NJ 07033
| | - Rubin Jiang
- Process Development and Engineering; Biologics and Vaccines, Merck & Co., Inc; 2000 Galloping Hill Road Kenilworth NJ 07033
| | - Hao Chen
- Process Development and Engineering; Biologics and Vaccines, Merck & Co., Inc; 2000 Galloping Hill Road Kenilworth NJ 07033
| |
Collapse
|