1
|
Shi Y, Wan Y, Yang J, Lu Y, Xie X, Pan J, Wang H, Qu H. Bioprocess biomarker identification and diagnosis for industrial mAb production based on metabolic profiling and multivariate data analysis. Bioprocess Biosyst Eng 2025; 48:771-783. [PMID: 40064687 DOI: 10.1007/s00449-025-03142-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025]
Abstract
Monoclonal antibody (mAb) production is a complex bioprocess influenced by various cellular and metabolic factors. Understanding these interactions is critical for optimizing manufacturing and improving yields. In this study, we proposed a diagnostic and identification strategy using quantitative proton nuclear magnetic resonance (1H qNMR) technology-based pharmaceutical process-omics to analyze bioprocess variability and unveil significant metabolites affecting cell growth and yield during industrial mAb manufacturing. First, batch level model (BLM) and orthogonal partial least squares-discriminant analysis (OPLS-DA) identified glucose and lactate as primary contributors to culture run variability. Maintaining an optimal glucose set point was crucial for high-yield runs. Second, a partial least squares (PLS) regression model was established, which revealed viable cell density (VCD), along with glutamine, maltose, tyrosine, citrate, methionine, and lactate, as critical variables impacting mAb yield. Finally, hierarchical clustering analysis (HCA) highlighted one-carbon metabolism metabolites, such as choline, pyroglutamate, and formate, as closely associated with VCD. These findings provide a foundation for future bioprocess optimization through cell line engineering and media formulation adjustments, ultimately enhancing mAb production efficiency.
Collapse
Affiliation(s)
- Yingting Shi
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuxiang Wan
- BioRay Pharmaceutical Co., Ltd, Taizhou, 318000, China
| | - Jiayu Yang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuting Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinyuan Xie
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianyang Pan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Haibin Wang
- BioRay Pharmaceutical Co., Ltd, Taizhou, 318000, China.
| | - Haibin Qu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Kienzle S, Junghans L, Wieschalka S, Diem K, Takors R, Radde NE, Kunzelmann M, Presser B, Nold V. Direct Consideration of Process History During Intensified Design of Experiments Planning Eases Interpretation of Mammalian Cell Culture Dynamics. Bioengineering (Basel) 2025; 12:319. [PMID: 40150783 PMCID: PMC11939677 DOI: 10.3390/bioengineering12030319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/26/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Intra-experimental factor setting shifts in intensified design of experiments (iDoE) enhance understanding of bioproduction processes by capturing their dynamics and are thus essential to fulfill quality by design (QbD) ambitions. Determining the influence of process history on the cellular responses, often referred to as memory effect, is fundamental for accurate predictions. However, the current iDoE designs do not explicitly consider nor quantify the influence of process history. Therefore, we propose the one-factor-multiple-columns (OFMC)-format for iDoE planning. This format explicitly describes stage-dependent factor effects and potential memory effects as across-stage interactions (ASIs) during a bioprocess. To illustrate its utility, an OFMC-iDoE that considers the characteristic growth phases during a fed-batch process was planned. Data were analyzed using ordinary least squares (OLS) regression as previously described via stage-wise analysis of the time series and compared to direct modeling of end-of-process outcomes enabled by the OFMC-format. This article aims to provide the reader with a framework on how to plan and model iDoE data and highlights how the OFMC-format simplifies planning, and data acquisition, eases modeling and gives a straightforward quantification of potential memory effects. With the proposed OFMC-format, optimization of bioprocesses can leverage which factor settings are most beneficial in which state of the mammalian culture and thus elevate performance and quality to the next level.
Collapse
Affiliation(s)
- Samuel Kienzle
- Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstraße 65, 88397 Biberach an der Riß, Germany
| | - Lisa Junghans
- Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstraße 65, 88397 Biberach an der Riß, Germany
| | - Stefan Wieschalka
- Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstraße 65, 88397 Biberach an der Riß, Germany
| | - Katharina Diem
- Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstraße 65, 88397 Biberach an der Riß, Germany
| | - Ralf Takors
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Nicole Erika Radde
- Institute for Stochastics and Applications, University of Stuttgart, Wankelstr. 5, 70563 Stuttgart, Germany
| | - Marco Kunzelmann
- Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstraße 65, 88397 Biberach an der Riß, Germany
| | - Beate Presser
- Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstraße 65, 88397 Biberach an der Riß, Germany
| | - Verena Nold
- Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstraße 65, 88397 Biberach an der Riß, Germany
- Global Computational Biology and Data Science, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstraße 65, 88397 Biberach an der Riß, Germany
| |
Collapse
|
3
|
Fu Y, Han Z, Cheng W, Niu S, Wang T, Wang X. Improvement strategies for transient gene expression in mammalian cells. Appl Microbiol Biotechnol 2024; 108:480. [PMID: 39365308 PMCID: PMC11452495 DOI: 10.1007/s00253-024-13315-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
Mammalian cells are suitable hosts for producing recombinant therapeutic proteins, with Chinese hamster ovary (CHO) and human embryonic kidney 293 (HEK293) cells being the most commonly used cell lines. Mammalian cell expression system includes stable and transient gene expression (TGE) system, with the TGE system having the advantages of short cycles and simple operation. By optimizing the TGE system, the expression of recombinant proteins has been significantly improved. Here, the TGE system and the detailed and up-to-date improvement strategies of mammalian cells, including cell line, expression vector, culture media, culture processes, transfection conditions, and co-expression of helper genes, are reviewed. KEY POINTS: • Detailed improvement strategies of transient gene expression system of mammalian cells are reviewed • The composition of transient expression system of mammalian cell are summarized • Proposed optimization prospects for transient gene expression systems.
Collapse
Affiliation(s)
- Yushun Fu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zimeng Han
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Wanting Cheng
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shuaichen Niu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tianyun Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Xiaoyin Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
4
|
Ghobadian H, Roshanzamir K, Mohammadhasan KA, Ostadi H, Zati Keikha R, Dolatkhah Baghan M, Talebkhan Y, Torkashvand F. Optimization of Culture Conditions to Improve Follicle-Stimulating Hormone Production by CHO-DG44 Cells in Serum-Free Medium. IRANIAN BIOMEDICAL JOURNAL 2024; 28:282-96. [PMID: 39901854 PMCID: PMC11829159 DOI: 10.61186/ibj.4160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/27/2024] [Indexed: 02/05/2025]
Abstract
Background In the present study, we attempted to adapt an adherent and serum-dependent Chinese hamster ovary DG44 cell line to a serum-free suspension culture and optimize the culture condition to achieve a higher yield of recombinant human follicle stimulating hormone (r-hFSH) with acceptable quality. This approach helps to mitigate the risks associated with blood-borne pathogens, reduces lot-to-lot variability, and lowers costs, making it suitable for industrial processing and scale-up. Methods The cell adaptation was performed using different chemically defined SFM. This process was followed by optimization through statistical experimental design, focusing on selected physicochemical parameters, including chemical supplementation of the medium and temperature shift. Both small- and large-scale cultures were conducted to test the reproducibility of the optimized condition. The expressed protein was evaluated for comparability with the standard molecule according to the Pharmacopeia guidelines. Results response surface methodology (RSM) analysis indicated that supplementation of the culture medium with galactose and sodium butyrate (NaBu), along with a temperature downshift, were the main parameters leading to increased cell viability (10%), r-hFSH level (96%), and more importantly, the glycosylation content (49%) of r-hFSH compared to the control condition. As r-hFSH isoforms generated during in vivo post-translational modifications typically exhibit different serum/plasma half-lives and bioactivity due to their incorporated sialic acid content/glycosylation, further optimizations of r-hFSH production are necessary to enhance its biological activity. In this study, following a primary screening of the studied parameters, optimization of culture conditions based on selected parameters resulted in enhanced quality and quantity of the produced r-hFSH. However, further examination is necessary before transitioning to industrial production.
Collapse
Affiliation(s)
- Hanna Ghobadian
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Khashayar Roshanzamir
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Kouhi Abdolabadi Mohammadhasan
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Hadi Ostadi
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Reza Zati Keikha
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Mohammad Dolatkhah Baghan
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Yeganeh Talebkhan
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
5
|
Rives D, Peak C, Blenner MA. RNASeq highlights ATF6 pathway regulators for CHO cell engineering with different impacts of ATF6β and WFS1 knockdown on fed-batch production of IgG 1. Sci Rep 2024; 14:14141. [PMID: 38898154 PMCID: PMC11187196 DOI: 10.1038/s41598-024-64767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
Secretion levels required of industrial Chinese hamster ovary (CHO) cell lines can challenge endoplasmic reticulum (ER) homeostasis, and ER stress caused by accumulation of misfolded proteins can be a bottleneck in biomanufacturing. The unfolded protein response (UPR) is initiated to restore homeostasis in response to ER stress, and optimization of the UPR can improve CHO cell production of therapeutic proteins. We compared the fed-batch growth, production characteristics, and transcriptomic response of an immunoglobulin G1 (IgG1) producer to its parental, non-producing host cell line. We conducted differential gene expression analysis using high throughput RNA sequencing (RNASeq) and quantitative polymerase chain reaction (qPCR) to study the ER stress response of each cell line during fed-batch culture. The UPR was activated in the IgG1 producer compared to the host cell line and our analysis of differential expression profiles indicated transient upregulation of ATF6α target mRNAs in the IgG1 producer, suggesting two upstream regulators of the ATF6 arm of the UPR, ATF6β and WFS1, are rational engineering targets. Although both ATF6β and WFS1 have been reported to negatively regulate ATF6α, this study shows knockdown of either target elicits different effects in an IgG1-producing CHO cell line. Stable knockdown of ATF6β decreased cell growth without decreasing titer; however, knockdown of WFS1 decreased titer without affecting growth. Relative expression measured by qPCR indicated no direct relationship between ATF6β and WFS1 expression, but upregulation of WFS1 in one pool was correlated with decreased growth and upregulation of ER chaperone mRNAs. While knockdown of WFS1 had negative impacts on UPR activation and product mRNA expression, knockdown of ATF6β improved the UPR specifically later in fed-batch leading to increased overall productivity.
Collapse
Affiliation(s)
- Dyllan Rives
- Department of Chemical & Biomolecular Engineering, Clemson University, 206 S. Palmetto Blvd., Clemson, SC, 29634-0909, USA
| | - Caroline Peak
- Department of Chemical & Biomolecular Engineering, Clemson University, 206 S. Palmetto Blvd., Clemson, SC, 29634-0909, USA
| | - Mark A Blenner
- Department of Chemical & Biomolecular Engineering, Clemson University, 206 S. Palmetto Blvd., Clemson, SC, 29634-0909, USA.
- Department of Chemical & Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, DE, 19713, USA.
| |
Collapse
|
6
|
González-Hernández Y, Perré P. Building blocks needed for mechanistic modeling of bioprocesses: A critical review based on protein production by CHO cells. Metab Eng Commun 2024; 18:e00232. [PMID: 38501051 PMCID: PMC10945193 DOI: 10.1016/j.mec.2024.e00232] [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: 10/25/2023] [Revised: 02/12/2024] [Accepted: 02/23/2024] [Indexed: 03/20/2024] Open
Abstract
This paper reviews the key building blocks needed to develop a mechanistic model for use as an operational production tool. The Chinese Hamster Ovary (CHO) cell, one of the most widely used hosts for antibody production in the pharmaceutical industry, is considered as a case study. CHO cell metabolism is characterized by two main phases, exponential growth followed by a stationary phase with strong protein production. This process presents an appropriate degree of complexity to outline the modeling strategy. The paper is organized into four main steps: (1) CHO systems and data collection; (2) metabolic analysis; (3) formulation of the mathematical model; and finally, (4) numerical solution, calibration, and validation. The overall approach can build a predictive model of target variables. According to the literature, one of the main current modeling challenges lies in understanding and predicting the spontaneous metabolic shift. Possible candidates for the trigger of the metabolic shift include the concentration of lactate and carbon dioxide. In our opinion, ammonium, which is also an inhibiting product, should be further investigated. Finally, the expected progress in the emerging field of hybrid modeling, which combines the best of mechanistic modeling and machine learning, is presented as a fascinating breakthrough. Note that the modeling strategy discussed here is a general framework that can be applied to any bioprocess.
Collapse
Affiliation(s)
- Yusmel González-Hernández
- Université Paris-Saclay, CentraleSupélec, Laboratoire de Génie des Procédés et Matériaux, Centre Européen de Biotechnologie et de Bioéconomie (CEBB), 3 Rue des Rouges Terres, 51110, Pomacle, France
| | - Patrick Perré
- Université Paris-Saclay, CentraleSupélec, Laboratoire de Génie des Procédés et Matériaux, Centre Européen de Biotechnologie et de Bioéconomie (CEBB), 3 Rue des Rouges Terres, 51110, Pomacle, France
| |
Collapse
|
7
|
Zhang X, Wang Y, Yi D, Zhang C, Ning B, Fu Y, Jia Y, Wang T, Wang X. Synergistic promotion of transient transgene expression in CHO cells by PDI/XBP-1s co-transfection and mild hypothermia. Bioprocess Biosyst Eng 2024; 47:557-565. [PMID: 38416261 DOI: 10.1007/s00449-024-02987-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 02/16/2024] [Indexed: 02/29/2024]
Abstract
Transient gene expression system is an important tool for rapid production of recombinant proteins in Chinese hamster ovary (CHO) cells. However, their low productivity is the main hurdle to overcome. An effective approach through which to obtain high protein yield involves targeting transcriptional, post-transcriptional events (PTEs), and culture conditions. Here, we investigated the effects of protein disulfide isomerase (PDI) and spliced X-box binding protein 1 (XBP-1s) co-overexpression combined with mild hypothermia on the transient yields of recombinant proteins in CHO cells. The results showed that the gene of interest (GOI) and the PDI/XBP-1s helper vector at a co-transfection ratio of 10:1 could obviously increase transient expression level of recombinant protein in CHO cells. However, PDI/XBP-1s overexpression had no significance effect on the mRNA levels of the recombinant protein, suggesting that it targeted PTEs. Moreover, the increased production was due to the enhancing of cell specific productivity, not related to cell growth, viability, and cell cycle. In addition, combined PDI/XBP-1s co-overexpression and mild hypothermia could further improve Adalimumab expression, compared to the control/37 °C and PDI/XBP-1s/37 °C, the Adalimumab volume yield of PDI/XBP-1s/33 °C increased by 203% and 142%, respectively. Mild hypothermia resulted in 3.52- and 2.33-fold increase in the relative mRNA levels of PDI and XBP-1s, respectively. In conclusion, the combination of PDI/XBP-1s overexpression and culture temperature optimization can achieve higher transient expression of recombinant protein, which provides a synergetic strategy to improve transient production of recombinant protein in CHO cells.
Collapse
Affiliation(s)
- Xi Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yaokun Wang
- The School of Medical Humanities, Xinxiang Medical University, Xinxiang, 453003, China
| | - Dandan Yi
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chi Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Binhuan Ning
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yushun Fu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yanlong Jia
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tianyun Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Xiaoyin Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
8
|
Geng SL, Zhao XJ, Zhang X, Zhang JH, Mi CL, Wang TY. Recombinant therapeutic proteins degradation and overcoming strategies in CHO cells. Appl Microbiol Biotechnol 2024; 108:182. [PMID: 38285115 PMCID: PMC10824870 DOI: 10.1007/s00253-024-13008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/20/2023] [Accepted: 01/08/2024] [Indexed: 01/30/2024]
Abstract
Mammalian cell lines are frequently used as the preferred host cells for producing recombinant therapeutic proteins (RTPs) having post-translational modified modification similar to those observed in proteins produced by human cells. Nowadays, most RTPs approved for marketing are produced in Chinese hamster ovary (CHO) cells. Recombinant therapeutic antibodies are among the most important and promising RTPs for biomedical applications. One of the issues that occurs during development of RTPs is their degradation, which caused by a variety of factors and reducing quality of RTPs. RTP degradation is especially concerning as they could result in reduced biological functions (antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity) and generate potentially immunogenic species. Therefore, the mechanisms underlying RTP degradation and strategies for avoiding degradation have regained an interest from academia and industry. In this review, we outline recent progress in this field, with a focus on factors that cause degradation during RTP production and the development of strategies for overcoming RTP degradation. KEY POINTS: • The recombinant therapeutic protein degradation in CHO cell systems is reviewed. • Enzymatic factors and non-enzymatic methods influence recombinant therapeutic protein degradation. • Reducing the degradation can improve the quality of recombinant therapeutic proteins.
Collapse
Affiliation(s)
- Shao-Lei Geng
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xiao-Jie Zhao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xi Zhang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Ji-Hong Zhang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Chun-Liu Mi
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Tian-Yun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| |
Collapse
|
9
|
Mao L, Schneider JW, Robinson AS. Rosmarinic acid enhances CHO cell productivity and proliferation through activation of the unfolded protein response and the mTOR pathway. Biotechnol J 2024; 19:e2300397. [PMID: 37897814 DOI: 10.1002/biot.202300397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/10/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
Rosmarinic acid (RA) has gained attraction in bioprocessing as a media supplement to improve cellular proliferation and protein production. Here, we observe up to a two-fold increase in antibody production with RA-supplementation, and a concentration-dependent effect of RA on cell proliferation for fed-batch Chinese hamster ovary (CHO) cell cultures. Contrary to previously reported antioxidant activity, RA increased the reactive oxygen species (ROS) levels, stimulated endoplasmic reticulum (ER) stress, activated the unfolded protein response (UPR), and elicited DNA damage. Despite such stressful events, RA appeared to maintained cell health via mammalian target of rapamycin (mTOR) pathway activation; both mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) were stimulated in RA-supplemented cultures. By reversing such mTOR pathway activity through either chemical inhibitor addition or siRNA knockdown of genes regulating the mTORC1 and mTORC2 complexes, antibody production, UPR signaling, and stress-induced DNA damage were reduced. Further, the proliferative effect of RA appeared to be regulated selectively by mTORC2 activation and have reproduced this observation by using the mTORC2 stimulator SC-79. Analogously, knockdown of mTORC2 strongly reduced X-box binding protein 1 (XBP1) splicing, which would be expected to reduce antibody folding and secretion, sugging that reduced mTORC2 would correlate with reduced antibody levels. The crosstalk between mTOR activation and UPR upregulation may thus be related directly to the enhanced productivity. Our results show the importance of the mTOR and UPR pathways in increasing antibody productivity, and suggest that RA supplementation may obviate the need for labor-intensive genetic engineering by directly activating pathways favorable to cell culture performance.
Collapse
Affiliation(s)
- Leran Mao
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - James W Schneider
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Anne S Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
Zhu Z, Chen X, Li W, Zhuang Y, Zhao Y, Wang G. Understanding the effect of temperature downshift on CHO cell growth, antibody titer and product quality by intracellular metabolite profiling and in vivo monitoring of redox state. Biotechnol Prog 2023; 39:e3352. [PMID: 37141532 DOI: 10.1002/btpr.3352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/16/2023] [Accepted: 04/20/2023] [Indexed: 05/06/2023]
Abstract
The strategy of temperature downshift has been widely used in the biopharmaceutical industry to improve antibody production and cell-specific production rate (qp ) with Chinese hamster ovary cells (CHO). However, the mechanism of temperature-induced metabolic rearrangement, especially important intracellular metabolic events, remains poorly understood. In this work, in order to explore the mechanisms of temperature-induced cell metabolism, we systematically assessed the differences in cell growth, antibody expression, and antibody quality between high-producing (HP) and low-producing (LP) CHO cell lines under both constant temperature (37°C) and temperature downshift (37°C→33°C) settings during fed-batch culture. Although the results showed that low-temperature culture during the late phase of exponential cell growth significantly reduced the maximum viable cell density (p < 0.05) and induced cell cycle arrest in the G0/G1 phase, this temperature downshift led to a higher cellular viability and increased antibody titer by 48% and 28% in HP and LP CHO cell cultures, respectively (p < 0.001), and favored antibody quality reflected in reduced charge heterogeneity and molecular size heterogeneity. Combined extra- and intra-cellular metabolomics analyses revealed that temperature downshift significantly downregulated intracellular glycolytic and lipid metabolic pathways while upregulated tricarboxylic acid (TCA) cycle, and particularly featured upregulated glutathione metabolic pathways. Interestingly, all these metabolic pathways were closely associated with the maintenance of intracellular redox state and oxidative stress-alleviating strategies. To experimentally address this, we developed two high-performance fluorescent biosensors, denoted SoNar and iNap1, for real-time monitoring of intracellular nicotinamide adenine dinucleotide/nicotinamide adenine dinucleotide + hydrogen (NAD+ /NADH) ratio and nicotinamide adenine dinucleotide phosphate (NADPH) amount, respectively. Consistent with such metabolic rearrangements, the results showed that temperature downshift decreased the intracellular NAD+ /NADH ratio, which might be ascribed to the re-consumption of lactate, and increased the intracellular NADPH amount (p < 0.01) to scavenge intracellular reactive oxygen species (ROS) induced by the increased metabolic requirements for high-level expression of antibody. Collectively, this study provides a metabolic map of cellular metabolic rearrangement induced by temperature downshift and demonstrates the feasibility of real-time fluorescent biosensors for biological processes, thus potentially providing a new strategy for dynamic optimization of antibody production processes.
Collapse
Affiliation(s)
- Ziyu Zhu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology (ECUST), Shanghai, China
| | - Xiaoqian Chen
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology (ECUST), Shanghai, China
| | - Wenhao Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology (ECUST), Shanghai, China
| | - Yingping Zhuang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology (ECUST), Shanghai, China
- Qingdao Innovation Institute of East China University of Science and Technology, Shanghai, China
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology (ECUST), Shanghai, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, China
| | - Guan Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology (ECUST), Shanghai, China
- Qingdao Innovation Institute of East China University of Science and Technology, Shanghai, China
| |
Collapse
|
11
|
Billerhart M, Hunjadi M, Hawlin V, Grünwald-Gruber C, Maresch D, Mayrhofer P, Kunert R. Recombinant Human CD19 in CHO-K1 Cells: Glycosylation Patterns as a Quality Attribute of High Yield Processes. Int J Mol Sci 2023; 24:10891. [PMID: 37446069 PMCID: PMC10341778 DOI: 10.3390/ijms241310891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
CD19 is an essential protein in personalized CD19-targeting chimeric antigen receptor (CAR)-T cell-based cancer immunotherapies and CAR-T cell functionality evaluation. However, the recombinant expression of this "difficult to-express" (DTE) protein is challenging, and therefore, commercial access to the protein is limited. We have previously described the successful stable expression of our soluble CD19-AD2 fusion protein of the CD19 extracellular part fused with human serum albumin domain 2 (AD2) in CHO-K1 cells. The function, stability, and secretion rate of DTE proteins can be improved by culture conditions, such as reduced temperature and a shorter residence time. Moreover, glycosylation, as one of the most important post-translational modifications, represents a critical quality attribute potentially affecting CAR-T cell effector function and thus impacting therapy's success. In this study, we increased the production rate of CD19-AD2 by 3.5-fold through applying hypothermic culture conditions. We efficiently improved the purification of our his-tagged CD19-AD2 fusion protein via a Ni-NTA-based affinity column using a stepwise increase in the imidazole concentration. The binding affinity to commercially available anti-CD19 antibodies was evaluated via Bio-Layer Interferometry (BLI). Furthermore, we revealed glycosylation patterns via Electrospray Ionization Mass Spectrometry (ESI-MS), and five highly sialylated and multi-antennary N-glycosylation sites were identified. In summary, we optimized the CD19-AD2 production and purification process and were the first to characterize five highly complex N-glycosylation sites.
Collapse
Affiliation(s)
- Magdalena Billerhart
- Institute of Animal Cell Technology and Systems Biology, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; (M.B.); (M.H.)
| | - Monika Hunjadi
- Institute of Animal Cell Technology and Systems Biology, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; (M.B.); (M.H.)
| | - Vanessa Hawlin
- Institute of Animal Cell Technology and Systems Biology, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; (M.B.); (M.H.)
| | - Clemens Grünwald-Gruber
- BOKU Core Facility Mass Spectrometry, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (C.G.-G.)
| | - Daniel Maresch
- BOKU Core Facility Mass Spectrometry, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (C.G.-G.)
| | - Patrick Mayrhofer
- Institute of Animal Cell Technology and Systems Biology, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; (M.B.); (M.H.)
| | - Renate Kunert
- Institute of Animal Cell Technology and Systems Biology, Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria; (M.B.); (M.H.)
| |
Collapse
|
12
|
Engineering of Chinese hamster ovary cells for co-overexpressing MYC and XBP1s increased cell proliferation and recombinant EPO production. Sci Rep 2023; 13:1482. [PMID: 36707606 PMCID: PMC9883479 DOI: 10.1038/s41598-023-28622-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/20/2023] [Indexed: 01/29/2023] Open
Abstract
Improving the cellular capacity of Chinese hamster ovary (CHO) cells to produce large amounts of therapeutic proteins remains a major challenge for the biopharmaceutical industry. In previous studies, we observed strong correlations between the performance of CHO cells and expression of two transcription factors (TFs), MYC and XBP1s. Here, we have evaluated the effective of overexpression of these two TFs on CHO cell productivity. To address this goal, we generated an EPO-producing cell line (CHOEPO) using a targeted integration approach, and subsequently engineered it to co-overexpress MYC and XBP1s (a cell line referred to as CHOCXEPO). Cells overexpressing MYC and XBP1s increased simultaneously viable cell densities and EPO production, leading to an enhanced overall performance in cultures. These improvements resulted from the individual effect of each TF in the cell behaviour (i.e., MYC-growth and XBP1s-productivity). An evaluation of the CHOCXEPO cells under different environmental conditions (temperature and media glucose concentration) indicated that CHOCXEPO cells increased cell productivity in high glucose concentration. This study showed the potential of combining TF-based cell engineering and process optimisation for increasing CHO cell productivity.
Collapse
|
13
|
Bartolo-Aguilar Y, Chávez-Cabrera C, Flores-Cotera LB, Badillo-Corona JA, Oliver-Salvador C, Marsch R. The potential of cold-shock promoters for the expression of recombinant proteins in microbes and mammalian cells. J Genet Eng Biotechnol 2022; 20:173. [PMID: 36580173 PMCID: PMC9800685 DOI: 10.1186/s43141-022-00455-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Low-temperature expression of recombinant proteins may be advantageous to support their proper folding and preserve bioactivity. The generation of expression vectors regulated under cold conditions can improve the expression of some target proteins that are difficult to express in different expression systems. The cspA encodes the major cold-shock protein from Escherichia coli (CspA). The promoter of cspA has been widely used to develop cold shock-inducible expression platforms in E. coli. Moreover, it is often necessary to employ expression systems other than bacteria, particularly when recombinant proteins require complex post-translational modifications. Currently, there are no commercial platforms available for expressing target genes by cold shock in eukaryotic cells. Consequently, genetic elements that respond to cold shock offer the possibility of developing novel cold-inducible expression platforms, particularly suitable for yeasts, and mammalian cells. CONCLUSIONS This review covers the importance of the cellular response to low temperatures and the prospective use of cold-sensitive promoters to direct the expression of recombinant proteins. This concept may contribute to renewing interest in applying white technologies to produce recombinant proteins that are difficult to express.
Collapse
Affiliation(s)
- Yaneth Bartolo-Aguilar
- Department of Biotechnology and Bioengineering, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, 07360, Mexico City, Mexico
- Instituto Politécnico Nacional-Unidad Profesional Interdisciplinaria de Biotecnología, Av. Acueducto s/n, Colonia Barrio La Laguna Ticomán, 07340, Mexico City, Mexico
| | - Cipriano Chávez-Cabrera
- Colegio de Estudios Científicos y Tecnológicos del Estado de Michoacán, CECyTE Michoacán, Héroes de la Revolución S/N, Col. Centro, 61880, Churumuco de Morelos, Michoacán, Mexico.
| | - Luis Bernardo Flores-Cotera
- Department of Biotechnology and Bioengineering, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, 07360, Mexico City, Mexico
| | - Jesús Agustín Badillo-Corona
- Instituto Politécnico Nacional-Unidad Profesional Interdisciplinaria de Biotecnología, Av. Acueducto s/n, Colonia Barrio La Laguna Ticomán, 07340, Mexico City, Mexico
| | - Carmen Oliver-Salvador
- Instituto Politécnico Nacional-Unidad Profesional Interdisciplinaria de Biotecnología, Av. Acueducto s/n, Colonia Barrio La Laguna Ticomán, 07340, Mexico City, Mexico
| | - Rodolfo Marsch
- Department of Biotechnology and Bioengineering, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, 07360, Mexico City, Mexico
| |
Collapse
|
14
|
Adjustable Thermo-Responsive, Cell-Adhesive Tissue Engineering Scaffolds for Cell Stimulation through Periodic Changes in Culture Temperature. Int J Mol Sci 2022; 24:ijms24010572. [PMID: 36614014 PMCID: PMC9820143 DOI: 10.3390/ijms24010572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
A three-dimensional (3D) scaffold ideally provides hierarchical complexity and imitates the chemistry and mechanical properties of the natural cell environment. Here, we report on a stimuli-responsive photo-cross-linkable resin formulation for the fabrication of scaffolds by continuous digital light processing (cDLP), which allows for the mechano-stimulation of adherent cells. The resin comprises a network-forming trifunctional acrylate ester monomer (trimethylolpropane triacrylate, or TMPTA), N-isopropyl acrylamide (NiPAAm), cationic dimethylaminoethyl acrylate (DMAEA) for enhanced cell interaction, and 4-acryloyl morpholine (AMO) to adjust the phase transition temperature (Ttrans) of the equilibrium swollen cross-polymerized scaffold. With glycofurol as a biocompatible solvent, controlled three-dimensional structures were fabricated and the transition temperatures were adjusted by resin composition. The effects of the thermally induced mechano-stimulation were investigated with mouse fibroblasts (L929) and myoblasts (C2C12) on printed constructs. Periodic changes in the culture temperature stimulated the myoblast proliferation.
Collapse
|
15
|
Enhanced recombinant protein production in CHO cell continuous cultures under growth-inhibiting conditions is associated with an arrested cell cycle in G1/G0 phase. PLoS One 2022; 17:e0277620. [PMCID: PMC9662745 DOI: 10.1371/journal.pone.0277620] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
Low temperature and sodium butyrate (NaBu) are two of the most used productivity-enhancing strategies in CHO cell cultures during biopharmaceutical manufacturing. While these two approaches alter the balance in the reciprocal relationship between cell growth and productivity, we do not fully understand their mechanisms of action beyond a gross cell growth inhibition. Here, we used continuous culture to evaluate the differential effect of low temperature and NaBu supplementation on CHO cell performance and gene expression profile. We found that an increase in cell-productivity under growth-inhibiting conditions was associated with the arrest of cells in the G1/G0 phase. A transcriptome analysis revealed that the molecular mechanisms by which low temperature and NaBu arrested cell cycle in G1/G0 differed from each other through the deregulation of different cell cycle checkpoints and regulators. The individual transcriptome changes in pattern observed in response to low temperature and NaBu were retained when these two strategies were combined, leading to an additive effect in arresting the cell cycle in G1/G0 phase. The findings presented here offer novel molecular insights about the cell cycle regulation during the CHO cell bioprocessing and its implications for increased recombinant protein production. This data provides a background for engineering productivity-enhanced CHO cell lines for continuous manufacturing.
Collapse
|
16
|
Fuentes P, Torres MJ, Arancibia R, Aulestia F, Vergara M, Carrión F, Osses N, Altamirano C. Dynamic Culture of Mesenchymal Stromal/Stem Cell Spheroids and Secretion of Paracrine Factors. Front Bioeng Biotechnol 2022; 10:916229. [PMID: 36046670 PMCID: PMC9421039 DOI: 10.3389/fbioe.2022.916229] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, conditioned medium (CM) obtained from the culture of mesenchymal stromal/stem cells (MSCs) has been shown to effectively promote tissue repair and modulate the immune response in vitro and in different animal models, with potential for application in regenerative medicine. Using CM offers multiple advantages over the implantation of MSCs themselves: 1) simpler storage, transport, and preservation requirements, 2) avoidance of the inherent risks of cell transplantation, and 3) potential application as a ready-to-go biologic product. For these reasons, a large amount of MSCs research has focused on the characterization of the obtained CM, including soluble trophic factors and vesicles, preconditioning strategies for enhancing paracrine secretion, such as hypoxia, a three-dimensional (3D) environment, and biochemical stimuli, and potential clinical applications. In vitro preconditioning strategies can increase the viability, proliferation, and paracrine properties of MSCs and therefore improve the therapeutic potential of the cells and their derived products. Specifically, dynamic cultivation conditions, such as fluid flow and 3D aggregate culture, substantially impact cellular behaviour. Increased levels of growth factors and cytokines were observed in 3D cultures of MSC grown on orbital or rotatory shaking platforms, in stirred systems, such as spinner flasks or stirred tank reactors, and in microgravity bioreactors. However, only a few studies have established dynamic culture conditions and protocols for 3D aggregate cultivation of MSCs as a scalable and reproducible strategy for CM production. This review summarizes significant advances into the upstream processing, mainly the dynamic generation and cultivation of MSC aggregates, for de CM manufacture and focuses on the standardization of the soluble factor production.
Collapse
Affiliation(s)
- Paloma Fuentes
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - María José Torres
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Rodrigo Arancibia
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Cellus Biomédica, Parque Tecnológico de León, León, Spain
| | - Francisco Aulestia
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Cellus Biomédica, Parque Tecnológico de León, León, Spain
| | - Mauricio Vergara
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Flavio Carrión
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Nelson Osses
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- CREAS, Centro Regional de Estudios en Alimentos Saludables, Valparaíso, Chile
- *Correspondence: Claudia Altamirano,
| |
Collapse
|
17
|
Li ZM, Fan ZL, Wang XY, Wang TY. Factors Affecting the Expression of Recombinant Protein and Improvement Strategies in Chinese Hamster Ovary Cells. Front Bioeng Biotechnol 2022; 10:880155. [PMID: 35860329 PMCID: PMC9289362 DOI: 10.3389/fbioe.2022.880155] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/01/2022] [Indexed: 01/20/2023] Open
Abstract
Recombinant therapeutic proteins (RTPs) are important parts of biopharmaceuticals. Chinese hamster ovary cells (CHO) have become the main cell hosts for the production of most RTPs approved for marketing because of their high-density suspension growth characteristics, and similar human post-translational modification patterns et al. In recent years, many studies have been performed on CHO cell expression systems, and the yields and quality of recombinant protein expression have been greatly improved. However, the expression levels of some proteins are still low or even difficult-to express in CHO cells. It is urgent further to increase the yields and to express successfully the “difficult-to express” protein in CHO cells. The process of recombinant protein expression of is a complex, involving multiple steps such as transcription, translation, folding processing and secretion. In addition, the inherent characteristics of molecular will also affect the production of protein. Here, we reviewed the factors affecting the expression of recombinant protein and improvement strategies in CHO cells.
Collapse
Affiliation(s)
- Zheng-Mei Li
- 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
| | - Zhen-Lin Fan
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang, China
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, China
| | - Xiao-Yin Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang, China
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| | - Tian-Yun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang, China
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Tian-Yun Wang,
| |
Collapse
|
18
|
A comprehensive comparison of mixing and mass transfer in shake flasks and their relationship with MAb productivity of CHO cells. Bioprocess Biosyst Eng 2022; 45:1033-1045. [DOI: 10.1007/s00449-022-02722-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 03/14/2022] [Indexed: 11/26/2022]
|
19
|
Torres M, Dickson AJ. Combined gene and environmental engineering offers a synergetic strategy to enhance r-protein production in Chinese hamster ovary cells. Biotechnol Bioeng 2021; 119:550-565. [PMID: 34821376 DOI: 10.1002/bit.28000] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/08/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022]
Abstract
Environmental growth-inhibition conditions (GICs) have been used extensively for increasing cell-specific productivity (qP ) of Chinese hamster ovary (CHO) cells, with the most common being temperature downshift and sodium butyrate (NaBu) treatment. B lymphocyte-induced maturation protein-1 (BLIMP1) overexpression in CHO cells can also inhibit cell growth and increase product titers and qP . Given the similar responses, this study evaluated the individual and combined effects of BLIMP1 expression, low temperature, and NaBu treatment on culture performance, cell metabolism, and recombinant protein production of CHO cells. As expected, all three interventions decreased cell growth, arrested cells in G1/G0 cell cycle phase, and increased qP . However, CHO cells presented different responses when considering cell viability, recombinant gene expression, and cell metabolism that indicated differences in the molecular loci by which BLIMP1 and GICs generated higher productivities. Combinations of BLIMP1 expression and GICs acted synergistically to inhibit cell growth and maximize r-protein production, with the BLIMP1/NaBu condition leading to the most significant improvements in product titers and qP . This latter condition also proved to substantially increase product yields (up to 9.8 g immunoglobulin G1 [IgG1]/L and 2.2 g erythropoietin-Fc [EPO-Fc]/L) and qP (up to 179 pg/cell/day [pcd] for IgG1 and 30 pcd for EPO-Fc) in high-density perfusion cultures. These findings offered mechanistic insights about the productivity-enhancing effects of BLIMP1 and GICs, as well as their complementarity for generating highly productive processes.
Collapse
Affiliation(s)
- Mauro Torres
- Faculty of Science and Engineering, Manchester Institute of Biotechnology, University of Manchester, Manchester, UK.,Department of Chemical Engineering and Analytical Science, Biochemical and Bioprocess Engineering Group, University of Manchester, Manchester, UK
| | - Alan J Dickson
- Faculty of Science and Engineering, Manchester Institute of Biotechnology, University of Manchester, Manchester, UK.,Department of Chemical Engineering and Analytical Science, Biochemical and Bioprocess Engineering Group, University of Manchester, Manchester, UK
| |
Collapse
|
20
|
Torres M, Akhtar S, McKenzie EA, Dickson AJ. Temperature Down-Shift Modifies Expression of UPR-/ERAD-Related Genes and Enhances Production of a Chimeric Fusion Protein in CHO Cells. Biotechnol J 2021; 16:e2000081. [PMID: 32271992 PMCID: PMC11475507 DOI: 10.1002/biot.202000081] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/29/2020] [Indexed: 02/05/2023]
Abstract
Low culture temperature enhances the cell-specific productivity of Chinese hamster ovary (CHO) cells expressing varied recombinant (r-) proteins, but the mechanisms remain unclear. Regulation of unfolded protein response (UPR) pathway genes, such as transcriptional regulatory factors and endoplasmic reticulum (ER)-resident proteins, appear to be involved in the improvements of r-protein production under low temperature conditions. The transcriptional regulation of UPR-specific targets is studied in response to decreased culture temperature in relation to production of a difficult-to-express protein. A clonally-derived CHO cell line expressing a chimeric fusion protein (human erythropoietin [hEPO] linked to a murine Fc region, hEPO-Fc) is evaluated in terms of growth, metabolism, r-protein production and UPR-/ER associated degradation (ERAD)-specific gene expression at standard (37 °C) and low (32 °C) temperature in batch and fed-batch systems. Low temperature decreased peak cell density, improved viability, generated cell cycle arrest in the G1 phase and enhanced hEPO-Fc expression in both batch and fed-batch cultures. A low culture temperature significantly upregulated genes encoding UPR-specific transcriptional activators (xbp1s, ddit3, and atf5) and ER-resident proteins (grp78, grp94, trib3, and ero1α), that are associated with folding and processing of proteins within the ER. Further, low culture temperature decreased expression of genes involved in ERAD (edem3, sels, herpud1, and syvn1) indicating a decreased potential for protein degradation.
Collapse
Affiliation(s)
- Mauro Torres
- Manchester Institute of BiotechnologyFaculty of Science and EngineeringUniversity of ManchesterManchesterM1 7DNUK
| | - Samia Akhtar
- Manchester Institute of BiotechnologyFaculty of Science and EngineeringUniversity of ManchesterManchesterM1 7DNUK
| | - Edward A. McKenzie
- Manchester Institute of BiotechnologyFaculty of Science and EngineeringUniversity of ManchesterManchesterM1 7DNUK
- Protein Expression FacilityManchester Institute of BiotechnologyFaculty of Life SciencesUniversity of ManchesterManchesterM1 7DNUK
| | - Alan J. Dickson
- Manchester Institute of BiotechnologyFaculty of Science and EngineeringUniversity of ManchesterManchesterM1 7DNUK
| |
Collapse
|
21
|
Torres M, Elvin M, Betts Z, Place S, Gaffney C, Dickson AJ. Metabolic profiling of Chinese hamster ovary cell cultures at different working volumes and agitation speeds using spin tube reactors. Biotechnol Prog 2020; 37:e3099. [PMID: 33169492 DOI: 10.1002/btpr.3099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/23/2020] [Accepted: 10/27/2020] [Indexed: 12/17/2022]
Abstract
Culture systems based on spin tube reactors have been consolidated in the development of manufacturing processes based on Chinese hamster ovary (CHO) cells. Despite their widespread use, there is little information about the consequences of varying operational setting parameters on the culture performance of recombinant CHO cell lines. Here, we investigated the effect of varying working volumes and agitation speeds on cell growth, protein production, and cell metabolism of two clonally derived CHO cell lines (expressing an IgG1 and a "difficult-to-express" fusion protein). Interestingly, low culture volumes increased recombinant protein production and decreased cell growth, while high culture volumes had the opposite effect. Altering agitation speeds exacerbated or moderated the differences observed due to culture volume changes. Combining low agitation rates with high culture volumes suppressed growth and recombinant protein production in CHO cells. Meanwhile, high agitation rates narrowed the differences in culture performance between low and high working volumes. These differences were also reflected in cell metabolism, where low culture volumes enhanced oxidative metabolism (linked to a productive phenotype) and high culture volume generated a metabolic profile that was predominately glycolytic (linked to a proliferative phenotype). Our findings indicate that the culture volume influence on metabolism modulates the balance between cell growth and protein production, a key feature that may be useful to adjust CHO cells toward a more productive phenotype.
Collapse
Affiliation(s)
- Mauro Torres
- Department of Chemical Engineering and Analytical Sciences, Faculty of Science & Engineering, Manchester Institute of Biotechnology, John Garside Building, The University of Manchester, Manchester, UK
| | - Mark Elvin
- Department of Chemical Engineering and Analytical Sciences, Faculty of Science & Engineering, Manchester Institute of Biotechnology, John Garside Building, The University of Manchester, Manchester, UK
| | - Zeynep Betts
- Department of Biology, Kocaeli University, Umuttepe Yerleskesi, Fen Edebiyat Fakultesi B Blok, Izmit, Turkey
| | - Svetlana Place
- Department of Chemical Engineering and Analytical Sciences, Faculty of Science & Engineering, Manchester Institute of Biotechnology, John Garside Building, The University of Manchester, Manchester, UK
| | - Claire Gaffney
- Department of Chemical Engineering and Analytical Sciences, Faculty of Science & Engineering, Manchester Institute of Biotechnology, John Garside Building, The University of Manchester, Manchester, UK
| | - Alan J Dickson
- Department of Chemical Engineering and Analytical Sciences, Faculty of Science & Engineering, Manchester Institute of Biotechnology, John Garside Building, The University of Manchester, Manchester, UK
| |
Collapse
|
22
|
Esposito D, Mehalko J, Drew M, Snead K, Wall V, Taylor T, Frank P, Denson JP, Hong M, Gulten G, Sadtler K, Messing S, Gillette W. Optimizing high-yield production of SARS-CoV-2 soluble spike trimers for serology assays. Protein Expr Purif 2020; 174:105686. [PMID: 32504802 PMCID: PMC7271859 DOI: 10.1016/j.pep.2020.105686] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 11/23/2022]
Abstract
The SARS-CoV-2 spike trimer is the primary antigen for several serology assays critical to determining the extent of SARS-CoV-2 exposure in the population. Until stable cell lines are developed to increase the titer of this secreted protein in mammalian cell culture, the low yield of spike protein produced from transient transfection of HEK293 cells will be a limiting factor for these assays. To improve the yield of spike protein and support the high demand for antigens in serology assays, we investigated several recombinant protein expression variables by altering the incubation temperature, harvest time, chromatography strategy, and final protein manipulation. Through this investigation, we developed a simplified and robust purification strategy that consistently yields 5 mg of protein per liter of expression culture for two commonly used forms of the SARS-CoV-2 spike protein. We show that these proteins form well-behaved stable trimers and are consistently functional in serology assays across multiple protein production lots.
Collapse
Affiliation(s)
- Dominic Esposito
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA.
| | - Jennifer Mehalko
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Matthew Drew
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Kelly Snead
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Vanessa Wall
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Troy Taylor
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Peter Frank
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - John-Paul Denson
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Min Hong
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Gulcin Gulten
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Kaitlyn Sadtler
- Section on Immuno-Engineering, National Institute for Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Simon Messing
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - William Gillette
- Protein Expression Laboratory, NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| |
Collapse
|
23
|
Esposito D, Mehalko J, Drew M, Snead K, Wall V, Taylor T, Frank P, Denson JP, Hong M, Gulten G, Sadtler K, Messing S, Gillette W. Optimizing high-yield production of SARS-CoV-2 soluble spike trimers for serology assays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.05.27.120204. [PMID: 32511418 PMCID: PMC7265690 DOI: 10.1101/2020.05.27.120204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The SARS-CoV-2 spike trimer is the primary antigen for several serology assays critical to determining the extent of SARS-CoV-2 exposure in the population. Until stable cell lines are developed to increase the titer of this secreted protein in mammalian cell culture, the low yield of spike protein produced from transient transfection of HEK293 cells will be a limiting factor for these assays. To improve the yield of spike protein and support the high demand for antigens in serology assays, we investigated several recombinant protein expression variables by altering the incubation temperature, harvest time, chromatography strategy, and final protein manipulation. Through this investigation, we developed a simplified and robust purification strategy that consistently yields 5 mg of protein per liter of expression culture for two commonly used forms of the SARS-CoV-2 spike protein. We show that these proteins form well-behaved stable trimers and are consistently functional in serology assays across multiple protein production lots.
Collapse
Affiliation(s)
- Dominic Esposito
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - Jennifer Mehalko
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - Matthew Drew
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - Kelly Snead
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - Vanessa Wall
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - Troy Taylor
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - Peter Frank
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - John-Paul Denson
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - Min Hong
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - Gulcin Gulten
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - Kaitlyn Sadtler
- Section on Immuno-Engineering, National Institute for Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda MD 20894
| | - Simon Messing
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| | - William Gillette
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. Frederick, MD 21702
| |
Collapse
|
24
|
McHugh KP, Xu J, Aron KL, Borys MC, Li ZJ. Effective temperature shift strategy development and scale confirmation for simultaneous optimization of protein productivity and quality in Chinese hamster ovary cells. Biotechnol Prog 2020; 36:e2959. [DOI: 10.1002/btpr.2959] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/17/2019] [Accepted: 01/06/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Kyle P. McHugh
- Global Product Development and SupplyBristol‐Myers Squibb Company Devens Massachusetts
| | - Jianlin Xu
- Global Product Development and SupplyBristol‐Myers Squibb Company Devens Massachusetts
| | - Kathryn L. Aron
- Global Product Development and SupplyBristol‐Myers Squibb Company Devens Massachusetts
| | - Michael C. Borys
- Global Product Development and SupplyBristol‐Myers Squibb Company Devens Massachusetts
| | - Zheng Jian Li
- Global Product Development and SupplyBristol‐Myers Squibb Company Devens Massachusetts
| |
Collapse
|
25
|
Torres M, Berrios J, Rigual Y, Latorre Y, Vergara M, Dickson AJ, Altamirano C. Metabolic flux analysis during galactose and lactate co-consumption reveals enhanced energy metabolism in continuous CHO cell cultures. Chem Eng Sci 2019. [DOI: 10.1016/j.ces.2019.04.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
26
|
Zúñiga RA, Gutiérrez-González M, Collazo N, Sotelo PH, Ribeiro CH, Altamirano C, Lorenzo C, Aguillón JC, Molina MC. Development of a new promoter to avoid the silencing of genes in the production of recombinant antibodies in chinese hamster ovary cells. J Biol Eng 2019; 13:59. [PMID: 31297150 PMCID: PMC6599231 DOI: 10.1186/s13036-019-0187-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/11/2019] [Indexed: 11/10/2022] Open
Abstract
Background The production of recombinant proteins in mammalian cell lines is one of the most important areas in biopharmaceutical industry. Viral transcriptional promoters are widely used to express recombinant proteins in mammalian cell lines. However, these promoters are susceptible to silencing, thus limiting protein productivity. Some CpG islands can avoid the silencing of housekeeping genes; for that reason, they have been used to increase the production of recombinant genes in cells of animal origin. In this study, we evaluated the CpG island of the promoter region of the β-actin gene of Cricetulus griseous (Chinese hamster), associated to the Cytomegalovirus (CMV) promoter, to increase recombinant antibodies production in Chinese Hamster Ovary (CHO) cells. Results We focused on the non-coding region of CpG island, which we called RegCG. RegCG behaved as a promoter, whose transcriptional activity was mainly commanded by the CAAT and CArG boxes of the proximal promoter. However, the transcription started mainly at the intronic region before the proximal transcription start site. While the CMV promoter was initially more powerful than RegCG, the latter promoter was more resistant to silencing than the CMV promoter in stable cell lines, and its activity was improved when combined with the CMV promoter. Thereby, the chimeric promoter was able to maintain the expression of recombinant antibodies in stable clones for 40 days at an average level 4 times higher than the CMV promoter. Finally, the chimeric promoter showed compatibility with a genetic amplification system by induction with methotrexate in cells deficient in the dihydrofolate reductase gene. Conclusions We have generated an efficient synthetic hybrid transcription promoter through the combination of RegCG with CMV, which, in stable cell lines, shows greater activity than when both promoters are used separately. Our chimeric promoter is compatible with a genetic amplification system in CHO DG44 cells and makes possible the generation of stable cell lines with high production of recombinant antibodies. We propose that this promoter can be a good alternative for the generation of clones expressing high amount of recombinant proteins, essential for industrial applications.
Collapse
Affiliation(s)
- Roberto A Zúñiga
- 1Centro de InmunoBiotecnología, Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,2Doctorado en Química, Universidad República Oriental del Uruguay, Montevideo, Uruguay
| | - Matías Gutiérrez-González
- 1Centro de InmunoBiotecnología, Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,7Programa de Doctorado en Farmacología, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Norberto Collazo
- 6Business Development Department, Fundación Fraunhofer Chile Research, Santiago, Chile
| | - Pablo Hérnan Sotelo
- 3Departamento de Biotecnología, Facultad de Ciencias Químicas, Universidad Nacional de Asunción, San Lorenzo, Paraguay
| | - Carolina H Ribeiro
- 1Centro de InmunoBiotecnología, Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Claudia Altamirano
- 5Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Carmen Lorenzo
- 4Facultad de Química, Universidad República Oriental del Uruguay, Montevideo, Uruguay
| | - Juan Carlos Aguillón
- 1Centro de InmunoBiotecnología, Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - María Carmen Molina
- 1Centro de InmunoBiotecnología, Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
27
|
Gutiérrez-González M, Latorre Y, Zúñiga R, Aguillón JC, Molina MC, Altamirano C. Transcription factor engineering in CHO cells for recombinant protein production. Crit Rev Biotechnol 2019; 39:665-679. [PMID: 31030575 DOI: 10.1080/07388551.2019.1605496] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The continuous increase of approved biopharmaceutical products drives the development of more efficient recombinant protein expression systems. Chinese hamster ovary (CHO) cells are the mainstay for this purpose but have some drawbacks, such as low levels of expression. Several strategies have been applied to increase the productivity of CHO cells with different outcomes. Transcription factor (TF) engineering has emerged as an interesting and successful approach, as these proteins can act as master regulators; the expression and function of a TF can be controlled by small molecules, and it is possible to design tailored TFs and promoters with desired features. To date, the majority of studies have focused on the use of TFs with growth, metabolic, cell cycle or endoplasmic reticulum functions, although there is a trend to develop new, synthetic TFs. Moreover, new synthetic biological approaches are showing promising advances for the development of specific TFs, even with tailored ligand sensitivity. In this article, we summarize the strategies to increase recombinant protein expression by modulating and designing TFs and with advancements in synthetic biology. We also illustrate how this class of proteins can be used to develop more robust expression systems.
Collapse
Affiliation(s)
| | - Yesenia Latorre
- b Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| | - Roberto Zúñiga
- a Centro de InmunoBiotecnología, Universidad de Chile , Santiago , Chile
| | | | | | - Claudia Altamirano
- b Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| |
Collapse
|
28
|
Xu J, Tang P, Yongky A, Drew B, Borys MC, Liu S, Li ZJ. Systematic development of temperature shift strategies for Chinese hamster ovary cells based on short duration cultures and kinetic modeling. MAbs 2019; 11:191-204. [PMID: 30230966 PMCID: PMC6343780 DOI: 10.1080/19420862.2018.1525262] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 09/02/2018] [Accepted: 09/13/2018] [Indexed: 10/28/2022] Open
Abstract
Temperature shift (TS) to a hypothermic condition has been widely used during protein production processes that use Chinese hamster ovary (CHO) cells. The effect of temperature on cell growth, metabolites, protein titer and quality depends on cell line, product, and other bioreactor conditions. Due to the large numbers of experiments, which typically last 2-3 weeks each, limited systematic TS studies have been reported with multiple shift temperatures and steps at different times. Here, we systematically studied the effect of temperature on cell culture performance for the production of two monoclonal antibodies by industrial GS and DG44 CHO cell lines. Three 2-8 day short-duration methods were developed and validated for researching the effect of many different temperatures on CHO cell culture and quality attributes. We found that minor temperature differences (1-1.5 °C) affected cell culture performance. The kinetic parameters extracted from the short duration data were subsequently used to compute and predict cell culture performance in extended duration of 10-14 days with multiple TS conditions for both CHO cell lines. These short-duration culture methods with kinetic modeling tools may be used for effective TS optimization to achieve the best profiles for cell growth, metabolites, titer and quality attributes. Although only three short-duration methods were developed with two CHO cell lines, similar short-duration methods with kinetic modeling may be applied for different hosts, including both microbial and other mammalian cells.
Collapse
Affiliation(s)
- Jianlin Xu
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| | - Peifeng Tang
- Department of Paper and Bioprocess Engineering, SUNY-ESF, Syracuse, NY, USA
| | - Andrew Yongky
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| | - Barry Drew
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| | - Michael C. Borys
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| | - Shijie Liu
- Department of Paper and Bioprocess Engineering, SUNY-ESF, Syracuse, NY, USA
| | - Zheng Jian Li
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| |
Collapse
|
29
|
Torres M, Altamirano C, Dickson AJ. Process and metabolic engineering perspectives of lactate production in mammalian cell cultures. Curr Opin Chem Eng 2018. [DOI: 10.1016/j.coche.2018.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
30
|
|