1
|
Lakshmanan M, Chia S, Pang KT, Sim LC, Teo G, Mak SY, Chen S, Lim HL, Lee AP, Bin Mahfut F, Ng SK, Yang Y, Soh A, Tan AHM, Choo A, Ho YS, Nguyen-Khuong T, Walsh I. Antibody glycan quality predicted from CHO cell culture media markers and machine learning. Comput Struct Biotechnol J 2024; 23:2497-2506. [PMID: 38966680 PMCID: PMC11222931 DOI: 10.1016/j.csbj.2024.05.046] [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: 02/19/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 07/06/2024] Open
Abstract
N-glycosylation can have a profound effect on the quality of mAb therapeutics. In biomanufacturing, one of the ways to influence N-glycosylation patterns is by altering the media used to grow mAb cell expression systems. Here, we explore the potential of machine learning (ML) to forecast the abundances of N-glycan types based on variables related to the growth media. The ML models exploit a dataset consisting of detailed glycomic characterisation of Anti-HER fed-batch bioreactor cell cultures measured daily under 12 different culture conditions, such as changes in levels of dissolved oxygen, pH, temperature, and the use of two different commercially available media. By performing spent media quantitation and subsequent calculation of pseudo cell consumption rates (termed media markers) as inputs to the ML model, we were able to demonstrate a small subset of media markers (18 selected out of 167 mass spectrometry peaks) in a Chinese Hamster Ovary (CHO) cell cultures are important to model N-glycan relative abundances (Regression - correlations between 0.80-0.92; Classification - AUC between 75.0-97.2). The performances suggest the ML models can infer N-glycan critical quality attributes from extracellular media as a proxy. Given its accuracy, we envisage its potential applications in biomaufactucuring, especially in areas of process development, downstream and upstream bioprocessing.
Collapse
Affiliation(s)
- Meiyappan Lakshmanan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, India
- Centre for Integrative Biology and Systems medicinE (IBSE), Indian Institute of Technology Madras, India
- Robert Bosch Centre for Data Science and AI (RBCDSAI), Indian Institute of Technology Madras, India
| | - Sean Chia
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Kuin Tian Pang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Lyn Chiin Sim
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Gavin Teo
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Shi Ya Mak
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Shuwen Chen
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Hsueh Lee Lim
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Alison P. Lee
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Farouq Bin Mahfut
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Say Kong Ng
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Annie Soh
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Andy Hee-Meng Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Andre Choo
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Ying Swan Ho
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Terry Nguyen-Khuong
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| | - Ian Walsh
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06–01 Centros, Singapore 138668, Republic of Singapore
| |
Collapse
|
2
|
Madabhushi SR, Chakravarty T, Kasza T, Padellan M, Atieh TB, Gupta B. Enhancing protein productivities in CHO cells through adenosine uptake modulation - Novel insights into cellular growth and productivity regulation. N Biotechnol 2024; 83:163-174. [PMID: 39151888 DOI: 10.1016/j.nbt.2024.08.500] [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: 05/21/2024] [Revised: 07/24/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Maximizing production potential of recombinant proteins such as monoclonal antibodies (mAbs) in Chinese Hamster Ovary (CHO) cells is a key enabler of reducing cost of goods of biologics. In this study, we explored various strategies to utilize adenosine mediated effects in biologics manufacturing processes. Results show that supplementation of adenosine increases specific productivity by up to two-fold while also arresting cell growth. Introducing adenosine in intensified perfusion processes in a biphasic manner significantly enhanced overall productivity. Interestingly, adenosine effect was observed to be dependent on the cell growth state. Using specific receptor antagonists and inhibitors, we identified that ENTs (primarily Slc29a1) mediate the uptake of adenosine in CHO cell cultures. Transcriptomics data showed an inverse correlation between Slc29a1 expression levels and peak viable cell densities. Data suggests that in fed-batch cultures, adenosine can be produced extracellularly. Blocking Slc29a1 using ENT inhibitors such as DZD and DP alone or in combination with CD73 inhibitor, PSB12379, resulted in a twofold increase in peak viable cell densities as well as productivities in fed batch - a novel strategy that can be applied to biologics manufacturing processes. This is the first study that suggests that adenosine production/accumulation in CHO cell cultures can potentially regulate the transition of CHO cells from exponential to stationary phase. We also demonstrate strategies to leverage this regulatory mechanism to maximize the productivity potential of biologics manufacturing processes.
Collapse
Affiliation(s)
| | | | - Tomas Kasza
- Biologics Process Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Malik Padellan
- Biologics Process Development, Merck & Co., Inc., Rahway, NJ, USA
| | | | - Balrina Gupta
- Biologics Process Development, Merck & Co., Inc., Rahway, NJ, USA
| |
Collapse
|
3
|
Cohrs M, Clottens N, Ramaut P, Braeckmans K, De Smedt S, Bauters T, Svilenov HL. Impact of pneumatic tube transportation on the aggregation of monoclonal antibodies in clinical practice. Eur J Pharm Sci 2024:106952. [PMID: 39481661 DOI: 10.1016/j.ejps.2024.106952] [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: 08/19/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Postproduction handling and in-hospital transportation of antibody drugs cause mechanical stress, including interfacial and shear stress, that can induce antibody unfolding and aggregation. The handling practices differ significantly between hospitals and the impact on protein stability is unknown. For example, the mechanical stress caused by transport via pneumatic tube systems (PTS) on therapeutic antibody aggregation is a potential safety and quality gap. The aim of this study was to investigate whether mechanical stress and PTS transportation in a hospital cause aggregation of five commonly used antibody drugs diluted in infusion bags. Orthogonal analytical methods showed that the handling and PTS transportation in this hospital did not cause aggregation of the investigated mAbs. The absence of aggregation could be explained by the reduction of interfacial stress due to headspace removal from the infusion bags and a mechanical sensor indicated that there was also only a moderate amount of mechanical stress caused by transportation with this particular PTS. Although this case study focuses on five antibody drugs and the practices in one hospital, the work demonstrates how to evaluate whether other handling and transportation practices cause significant mechanical stress that could compromise the quality and safety of antibody drugs.
Collapse
Affiliation(s)
- Michaela Cohrs
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Nele Clottens
- Pharmacy Department, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Pieter Ramaut
- Pharmacy Department, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Tiene Bauters
- Pharmacy Department, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium,.
| | - Hristo L Svilenov
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Biopharmaceutical Technology, TUM School of Life Sciences, Technical University of Munich, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany.
| |
Collapse
|
4
|
Lee HM, Kim TH, Park JH, Heo NY, Kim HS, Kim DE, Lee MK, Lee GM, You J, Kim YG. Sialyllactose supplementation enhances sialylation of Fc-fusion glycoprotein in recombinant Chinese hamster ovary cell culture. J Biotechnol 2024; 392:180-189. [PMID: 39038661 DOI: 10.1016/j.jbiotec.2024.07.016] [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: 04/03/2024] [Revised: 07/03/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Sialylation during N-glycosylation plays an important role in the half-life of therapeutic glycoproteins in vivo and has sparked interest in the production of therapeutic proteins using recombinant Chinese hamster ovary (rCHO) cells. To improve the sialylation of therapeutic proteins, we examined the effect of sialyllactose supplementation on sialylation of Fc-fusion glycoproteins produced in rCHO cells. Two enzymatically-synthesized sialyllactoses, 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL), were administered separately to two rCHO cell lines producing the same Fc-fusion glycoprotein derived from DUKX-B11 and DG44, respectively. Two sialyllactoses successfully increased sialylation of Fc-fusion glycoprotein in both cell lines, as evidenced by isoform distribution, sialylated N-glycan formation, and sialic acid content. Increased sialylation by adding sialyllactose was likely the result of increased amount of intracellular CMP-sialic acid (CMP-SA), the direct nucleotide sugar for sialylation. Furthermore, the degree of sialylation enhanced by sialyllactoses was slightly effective or nearly similar compared with the addition of N-acetylmannosamine (ManNAc), a representative nucleotide sugar precursor, to increase sialylation of glycoproteins. The effectiveness of sialyllactose was also confirmed using three commercially available CHO cell culture media. Taken together, these results suggest that enzymatically-synthesized sialyllactose represents a promising candidate for culture media supplementation to increase sialylation of glycoproteins in rCHO cell culture.
Collapse
Affiliation(s)
- Hoon-Min Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea; Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, South Korea
| | - Tae-Ho Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea; Department of Plant and Environmental New Resources, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, South Korea
| | - Jong-Ho Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea; Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, South Korea
| | - Na-Yeong Heo
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea; Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, South Korea
| | - Hyun-Seung Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea; Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, South Korea
| | - Dae Eung Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Mi Kyeong Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, South Korea
| | - Jungmok You
- Department of Plant and Environmental New Resources, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, South Korea.
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea; Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, South Korea.
| |
Collapse
|
5
|
Zhang J, Yang W, Zhang L, Li W, Zhang X, Wang X, Wang T. Novel and effective screening system for recombinant protein production in CHO cells. Sci Rep 2024; 14:20856. [PMID: 39242806 PMCID: PMC11379927 DOI: 10.1038/s41598-024-71915-0] [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: 05/01/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024] Open
Abstract
At present, biopharmaceuticals have received extensive attention from the society, among which recombinant proteins have a good growth trend and a large market share. Chinese hamster ovary (CHO) cells are the preferred mammalian system to produce glycosylated recombinant protein drugs. A highly efficient and stable cell screening method needs to be developed to obtain more and useful recombinant proteins. Limited dilution method, cell sorting, and semi-solid medium screening are currently the commonly used cell cloning methods. These methods are time-consuming and labor-intensive, and they have the disadvantage of low clone survival rate. Here, a method based on semi-solid medium was developed to screen out high-yielding and stable cell line within 3 weeks to improve the screening efficiency. The semi-solid medium was combined with an expression vector containing red fluorescent protein (RFP) for early cell line development. In accordance with the fluorescence intensity of RFP, the expression of upstream target gene could be indicated, and the fluorescence intensity was in direct proportion to the expression of upstream target gene. In conclusion, semi-solid medium combined with bicistronic expression vector provides an efficient method for screening stable and highly expressed cell lines.
Collapse
Affiliation(s)
- Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China.
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China.
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Wenwen Yang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liao Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| | - Wenqing Li
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xi Zhang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| | - Xiaoyin Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tianyun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China.
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
6
|
Liu Q, Hong J, Zhang Y, Wang Q, Xia Q, Knierman MD, Lau J, Dayaratna C, Negron B, Nanda H, Gunawardena HP. Rapid identification of antibody impurities in size-based electrophoresis via CZE-MS generated spectral library. Sci Rep 2024; 14:20239. [PMID: 39215123 PMCID: PMC11364755 DOI: 10.1038/s41598-024-70914-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Methods for the reliable and effective detection and identification of impurities are crucial to ensure the quality and safety of biopharmaceutical products. Technical limitations constrain the accurate identification of individual impurity peaks by size-based electrophoresis separations followed by mass spectrometry. This study presents a size-based electrophoretic method for detecting and identifying impurity peaks in antibody production. A hydrogen sulfide-accelerated degradation method was employed to generate known degradation products observed in bioreactors that forms the basis for size calibration. LabChip GXII channel electrophoresis enabled the rapid (< 1 min) detection of impurity peaks based on size, while capillary zone electrophoresis-mass spectrometry (CZE-MS) facilitated their accurate identification. We combine these techniques to examine impurities resulting from cell culture harvest conditions and forced degradation to assess antibody stability. To mimic cell culture harvest conditions and the impact of forced degradation, we subjected samples to cathepsin at different pH buffers or exposed them to high pH and temperature. Our method demonstrated the feasibility and broad applicability of using a CZE-MS generated spectral library to unambiguously assign peaks in high throughput size-based electrophoresis (i.e., LabChip GXII) with identifications or likely mass of the antibody impurity. Overall, this strategy combines the utility of CZE-MS as a high-resolution separation and detection method for impurities with size-based electrophoresis methods that are typically used to detect (not identify) impurities during the discovery and development of antibody therapeutics.
Collapse
Affiliation(s)
- Quan Liu
- CMP Scientific Corp, 760 Parkside Ave, STE 211, Brooklyn, NY, 11226, USA
| | - Jiaying Hong
- CMP Scientific Corp, 760 Parkside Ave, STE 211, Brooklyn, NY, 11226, USA
| | - Yukun Zhang
- CMP Scientific Corp, 760 Parkside Ave, STE 211, Brooklyn, NY, 11226, USA
| | - Qiuyue Wang
- CMP Scientific Corp, 760 Parkside Ave, STE 211, Brooklyn, NY, 11226, USA
| | - Qiangwei Xia
- CMP Scientific Corp, 760 Parkside Ave, STE 211, Brooklyn, NY, 11226, USA
| | - Michael D Knierman
- Agilent Technologies, 5301 Stevens Creek Blvd, Santa Clara, CA, 95051, USA
| | - Jim Lau
- Agilent Technologies, 5301 Stevens Creek Blvd, Santa Clara, CA, 95051, USA
| | - Caleen Dayaratna
- Johnson & Johnson Innovative Medicine Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Benjamin Negron
- Johnson & Johnson Innovative Medicine Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Hirsh Nanda
- Johnson & Johnson Innovative Medicine Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Harsha P Gunawardena
- Johnson & Johnson Innovative Medicine Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA.
| |
Collapse
|
7
|
Okamura K, Badr S, Ichida Y, Yamada A, Sugiyama H. Modeling of cell cultivation for monoclonal antibody production processes considering lactate metabolic shifts. Biotechnol Prog 2024:e3486. [PMID: 38924316 DOI: 10.1002/btpr.3486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/10/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Demand for monoclonal antibodies (mAbs) is rapidly increasing. To achieve higher productivity, there have been improvements to cell lines, operating modes, media, and cultivation conditions. Representative mathematical models are needed to narrow down the growing number of process alternatives. Previous studies have proposed mechanistic models to depict cell metabolic shifts (e.g., lactate production to consumption). However, the impacts of variations of some operating conditions have not yet been fully incorporated in such models. This paper offers a new mechanistic model considering variations in dissolved oxygen and glutamine depletion on cell metabolism applied to a novel Chinese hamster ovary (CHO) cell line. Expressions for the specific rates of lactate production, glutamine consumption, and mAb production were formulated for stirred and shaken-tank reactors. A deeper understanding of lactate metabolic shifts was obtained under different combinations of experimental conditions. Lactate consumption was more pronounced in conditions with higher DO and low glutamine concentrations. The model offers mechanistic insights that are useful for designing advanced operation strategies. It can be used in design space generation and process optimization for better productivity and product quality.
Collapse
Affiliation(s)
- Kozue Okamura
- Department of Chemical System Engineering, The University of Tokyo, Tokyo, Japan
| | - Sara Badr
- Department of Chemical System Engineering, The University of Tokyo, Tokyo, Japan
| | - Yusuke Ichida
- Department of Chemical System Engineering, The University of Tokyo, Tokyo, Japan
| | - Akira Yamada
- Department of Chemical System Engineering, The University of Tokyo, Tokyo, Japan
| | - Hirokazu Sugiyama
- Department of Chemical System Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Yamaguchi T, Ishikawa H, Fukuda M, Sugita Y, Furuie M, Nagano R, Suzawa T, Yamamoto K, Wakamatsu K. Catechins prevent monoclonal antibody fragmentation during production via fed-batch culture of Chinese hamster ovary cells. Biotechnol Prog 2024; 40:e3447. [PMID: 38415979 DOI: 10.1002/btpr.3447] [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: 12/10/2023] [Revised: 02/14/2024] [Accepted: 02/14/2024] [Indexed: 02/29/2024]
Abstract
Chinese hamster ovary (CHO) cells are widely used for the industrial production of therapeutic monoclonal antibodies (mAbs). To meet the increasing market demands, high productivity, and quality are required in cell culture. One of the critical attributes of mAbs, from a safety perspective, is mAb fragmentation. However, methods for preventing mAbs fragmentation in CHO cell culture are limited. In this study, we observed that the antibody fragment content increased with increasing titers in fed-batch cultures for all three cell lines expressing recombinant antibodies. Adding copper sulfate to the culture medium further increased the fragment content, suggesting the involvement of reactive oxygen species (ROS) in the fragmentation process. Though antioxidants may be helpful to scavenge ROS, several antioxidants are reported to decrease the productivity of CHO cells. Among the antioxidants examined, we observed that the addition of catechin or (-)-epigallocatechin gallate to the culture medium prevented fragmentation content by about 20% and increased viable cell density and titer by 30% and 10%, respectively. Thus, the addition of catechins or compounds of equivalent function would be beneficial for manufacturing therapeutic mAbs with a balance between high titers and good quality.
Collapse
Affiliation(s)
- Tsuyoshi Yamaguchi
- Graduate School of Science and Technology, Gunma University, Gunma, Japan
- Bio Process Research and Development Laboratories, Production Division, Kyowa Kirin Co. Ltd., Takasaki, Gunma, Japan
| | - Hiroko Ishikawa
- Bio Process Research and Development Laboratories, Production Division, Kyowa Kirin Co. Ltd., Takasaki, Gunma, Japan
| | - Mie Fukuda
- Bio Process Research and Development Laboratories, Production Division, Kyowa Kirin Co. Ltd., Takasaki, Gunma, Japan
| | - Yumi Sugita
- Bio Process Research and Development Laboratories, Production Division, Kyowa Kirin Co. Ltd., Takasaki, Gunma, Japan
| | - Misaki Furuie
- Bio Process Research and Development Laboratories, Production Division, Kyowa Kirin Co. Ltd., Takasaki, Gunma, Japan
| | - Ryuma Nagano
- Bio Process Research and Development Laboratories, Production Division, Kyowa Kirin Co. Ltd., Takasaki, Gunma, Japan
| | | | - Koichi Yamamoto
- Bio Process Research and Development Laboratories, Production Division, Kyowa Kirin Co. Ltd., Takasaki, Gunma, Japan
| | - Kaori Wakamatsu
- Graduate School of Science and Technology, Gunma University, Gunma, Japan
| |
Collapse
|
9
|
Lee HM, Park JH, Kim TH, Kim HS, Kim DE, Lee MK, You J, Lee GM, Kim YG. Effects of autophagy-inhibiting chemicals on sialylation of Fc-fusion glycoprotein in recombinant CHO cells. Appl Microbiol Biotechnol 2024; 108:224. [PMID: 38376550 PMCID: PMC10879319 DOI: 10.1007/s00253-024-13059-9] [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: 10/20/2023] [Revised: 01/27/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024]
Abstract
The occurrence of autophagy in recombinant Chinese hamster ovary (rCHO) cell culture has attracted attention due to its effects on therapeutic protein production. Given the significance of glycosylation in therapeutic proteins, this study examined the effects of autophagy-inhibiting chemicals on sialylation of Fc-fusion glycoproteins in rCHO cells. Three chemical autophagy inhibitors known to inhibit different stages were separately treated with two rCHO cell lines that produce the same Fc-fusion glycoprotein derived from DUKX-B11 and DG44. All autophagy inhibitors significantly decreased the sialylation of Fc-fusion glycoprotein in both cell lines. The decrease in sialylation of Fc-fusion glycoprotein is unlikely to be attributed to the release of intracellular enzymes, given the high cell viability and low activity of extracellular sialidases. Interestingly, the five intracellular nucleotide sugars remained abundant in cells treated with autophagy inhibitors. In the mRNA expression profiles of 27 N-glycosylation-related genes using the NanoString nCounter system, no significant differences in gene expression were noted. With the positive effect of supplementing nucleotide sugar precursors on sialylation, attempts were made to enhance the levels of intracellular nucleotide sugars by supplying these precursors. The addition of nucleotide sugar precursors to cultures treated with inhibitors successfully enhanced the sialylation of Fc-fusion glycoproteins compared to the control culture. This was particularly evident under mild stress conditions and not under relatively severe stress conditions, which were characterized by a high decrease in sialylation. These results suggest that inhibiting autophagy in rCHO cell culture decreases sialylation of Fc-fusion glycoprotein by constraining the availability of intracellular nucleotide sugars. KEY POINTS: • The autophagy inhibition in rCHO cell culture leads to a significant reduction in the sialylation of Fc-fusion glycoprotein. • The pool of five intracellular nucleotide sugars remained highly abundant in cells treated with autophagy inhibitors. • Supplementation of nucleotide sugar precursors effectively restores decreased sialylation, particularly under mild stress conditions but not in relatively severe stress conditions.
Collapse
Affiliation(s)
- Hoon-Min Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea
| | - Jong-Ho Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea
- Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, Korea
| | - Tae-Ho Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea
- Department of Plant and Environmental New Resources, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, Korea
| | - Hyun-Seung Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea
| | - Dae Eung Kim
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Korea
| | - Mi Kyeong Lee
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Korea
| | - Jungmok You
- Department of Plant and Environmental New Resources, Graduate School of Biotechnology, College of Life Science, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, 335 Gwahak-ro, Yuseong-gu, Daejeon, Korea
| | - Yeon-Gu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Korea.
- Department of Bioprocess Engineering, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea.
| |
Collapse
|
10
|
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
|
11
|
Wysor SK, Marcus RK. Quantitative Recoveries of Exosomes and Monoclonal Antibodies from Chinese Hamster Ovary Cell Cultures by Use of a Single, Integrated Two-Dimensional Liquid Chromatography Method. Anal Chem 2023; 95:17886-17893. [PMID: 37995145 PMCID: PMC11095952 DOI: 10.1021/acs.analchem.3c04044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
Cultured cell lines are very commonly used for the mass production of therapeutic proteins, such as monoclonal antibodies (mAbs). In particular, Chinese hamster ovary (CHO) cell lines are widely employed due to their high tolerance to variations in experimental conditions and their ability to grow in suspension or serum free media. CHO cell lines are known for their ability to produce high titers of biotherapeutic products such as immunoglobulin G (IgG). An emergent alternative means of treating diseases, such as cancer, is the use of gene therapies, wherein genetic cargo is "packaged" in nanosized vesicular structures, referred to as "vectors". One particularly attractive vector option is extracellular vesicles (EVs), of which exosomes are of greatest interest. While exosomes can be harvested from virtually any human body fluid, bovine milk, or even plants, their production in cell cultures is an attractive commercial approach. In fact, the same CHO cell types employed for mAb production also produce exosomes as a natural byproduct. Here, we describe a single integrated 2D liquid chromatography (2DLC) method for the quantitative recovery of both exosomes and antibodies from a singular sample aliquot. At the heart of the method is the use of polyester capillary-channeled polymer (C-CP) fibers as the first dimension column, wherein exosomes/EVs are captured from the supernatant sample and subsequently determined by multiangle light scattering (MALS), while the mAbs are captured, eluted, and quantified using a protein A-modified C-CP fiber column in the second dimension, all in a 10 min workflow. These efforts demonstrate the versatility of the C-CP fiber phases with the capacity to harvest both forms of therapeutics from a single bioreactor, suggesting an appreciable potential impact in the field of biotherapeutics production.
Collapse
Affiliation(s)
- Sarah K Wysor
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, South Carolina 29634-0973, United States
| | - R Kenneth Marcus
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, South Carolina 29634-0973, United States
| |
Collapse
|
12
|
Ying B, Kawabe Y, Zheng F, Amamoto Y, Kamihira M. High-Level Production of scFv-Fc Antibody Using an Artificial Promoter System with Transcriptional Positive Feedback Loop of Transactivator in CHO Cells. Cells 2023; 12:2638. [PMID: 37998372 PMCID: PMC10670205 DOI: 10.3390/cells12222638] [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: 10/17/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
With the increasing demand for therapeutic antibodies, CHO cells have become the de facto standard as producer host cells for biopharmaceutical production. High production yields are required for antibody production, and developing a high-titer production system is increasingly crucial. This study was established to develop a high-production system using a synthetic biology approach by designing a gene expression system based on an artificial transcription factor that can strongly induce the high expression of target genes in CHO cells. To demonstrate the functionality of this artificial gene expression system and its ability to induce the high expression of target genes in CHO cells, a model antibody (scFv-Fc) was produced using this system. Excellent results were obtained with the plate scale, and when attempting continuous production in semi-continuous cultures using bioreactor tubes with high-cell-density suspension culture using a serum-free medium, high-titer antibody production at the gram-per-liter level was achieved. Shifting the culture temperature to a low temperature of 33 °C achieved scFv-Fc concentrations of up to 5.5 g/L with a specific production rate of 262 pg/(cell∙day). This artificial gene expression system should be a powerful tool for CHO cell engineering aimed at constructing high-yield production systems.
Collapse
Affiliation(s)
| | | | | | | | - Masamichi Kamihira
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (B.Y.); (Y.K.); (F.Z.); (Y.A.)
| |
Collapse
|
13
|
Vitharana S, Stillahn JM, Katayama DS, Henry CS, Manning MC. Application of Formulation Principles to Stability Issues Encountered During Processing, Manufacturing, and Storage of Drug Substance and Drug Product Protein Therapeutics. J Pharm Sci 2023; 112:2724-2751. [PMID: 37572779 DOI: 10.1016/j.xphs.2023.08.003] [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: 10/14/2022] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
The field of formulation and stabilization of protein therapeutics has become rather extensive. However, most of the focus has been on stabilization of the final drug product. Yet, proteins experience stress and degradation through the manufacturing process, starting with fermentaition. This review describes how formulation principles can be applied to stabilize biopharmaceutical proteins during bioprocessing and manufacturing, considering each unit operation involved in prepration of the drug substance. In addition, the impact of the container on stabilty is discussed as well.
Collapse
Affiliation(s)
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
14
|
Reddy JV, Raudenbush K, Papoutsakis ET, Ierapetritou M. Cell-culture process optimization via model-based predictions of metabolism and protein glycosylation. Biotechnol Adv 2023; 67:108179. [PMID: 37257729 DOI: 10.1016/j.biotechadv.2023.108179] [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: 11/27/2022] [Revised: 05/18/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
In order to meet the rising demand for biologics and become competitive on the developing biosimilar market, there is a need for process intensification of biomanufacturing processes. Process development of biologics has historically relied on extensive experimentation to develop and optimize biopharmaceutical manufacturing. Experimentation to optimize media formulations, feeding schedules, bioreactor operations and bioreactor scale up is expensive, labor intensive and time consuming. Mathematical modeling frameworks have the potential to enable process intensification while reducing the experimental burden. This review focuses on mathematical modeling of cellular metabolism and N-linked glycosylation as applied to upstream manufacturing of biologics. We review developments in the field of modeling cellular metabolism of mammalian cells using kinetic and stoichiometric modeling frameworks along with their applications to simulate, optimize and improve mechanistic understanding of the process. Interest in modeling N-linked glycosylation has led to the creation of various types of parametric and non-parametric models. Most published studies on mammalian cell metabolism have performed experiments in shake flasks where the pH and dissolved oxygen cannot be controlled. Efforts to understand and model the effect of bioreactor-specific parameters such as pH, dissolved oxygen, temperature, and bioreactor heterogeneity are critically reviewed. Most modeling efforts have focused on the Chinese Hamster Ovary (CHO) cells, which are most commonly used to produce monoclonal antibodies (mAbs). However, these modeling approaches can be generalized and applied to any mammalian cell-based manufacturing platform. Current and potential future applications of these models for Vero cell-based vaccine manufacturing, CAR-T cell therapies, and viral vector manufacturing are also discussed. We offer specific recommendations for improving the applicability of these models to industrially relevant processes.
Collapse
Affiliation(s)
- Jayanth Venkatarama Reddy
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA
| | - Katherine Raudenbush
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA; Delaware Biotechnology Institute, Department of Biological Sciences, University of Delaware, USA.
| | - Marianthi Ierapetritou
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA.
| |
Collapse
|
15
|
Zhang D, Qiu J, Niu QT, Liu T, Gu R, Zhang X, Luo S. Effects of various pine needle extracts on Chinese hamster ovary cell growth and monoclonal antibody quality. Prep Biochem Biotechnol 2023; 53:1081-1091. [PMID: 36756987 DOI: 10.1080/10826068.2023.2166959] [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: 02/10/2023]
Abstract
Chinese hamster ovary (CHO) cells are commonly used as "bio-machines" to pro-duce monoclonal antibodies (mAb) because of their ability to produce very complex proteins. In this study, we evaluated the effects of pine needle water extract (PNWE), pine needle ethanol extract (PNEE), and pine needle polysaccharide extract (PNPE) on the CHO cell growth, mAb production and quality using a Fed-batch culture process. PNPE maintained high VCD and viability, and the titer increase was correlated with its concentration. Three extracts effectively reduced the acidic charge variant and modulated mAb glycosylation. PNPE had the most profound effect, with G0F decreasing by 8.7% and G1Fa increasing by 6.7%. The change in the glycoform was also closely related to the PNPE concentration. This study demonstrated that PNPE could facilitate CHO cell growth, increase the mAb production, decrease acidic charge variants, and regulate mAb glycoforms. To identify the components responsible for the above changes, the sugar and flavonoid contents in the extracts were determined, and the chemical compounds were identified by LC-MS, resulting in 38 compounds identified from PNPE. Rich in sugars and flavonoids in these three extracts may be related to increased CHO cell growth and productivity, and changes in glycoforms.
Collapse
Affiliation(s)
- Dingyue Zhang
- Anhui University of Chinese Medicine, Hefei, China
- Yangtze Delta Drug Advanced Research Institute, Nantong, China
| | - Jinshu Qiu
- Thousand Oaks Biopharmaceuticals Co., Ltd., Nantong, China
| | - Qing-Tian Niu
- Thousand Oaks Biopharmaceuticals Co., Ltd., Nantong, China
| | - Tingting Liu
- Thousand Oaks Biopharmaceuticals Co., Ltd., Nantong, China
| | - Rulin Gu
- Thousand Oaks Biopharmaceuticals Co., Ltd., Nantong, China
| | - Xiaoying Zhang
- Thousand Oaks Biopharmaceuticals Co., Ltd., Nantong, China
| | - Shun Luo
- Anhui University of Chinese Medicine, Hefei, China
- Thousand Oaks Biopharmaceuticals Co., Ltd., Nantong, China
| |
Collapse
|
16
|
Maltais JS, Lord-Dufour S, Morasse A, Stuible M, Loignon M, Durocher Y. Repressing expression of difficult-to-express recombinant proteins during the selection process increases productivity of CHO stable pools. Biotechnol Bioeng 2023; 120:2840-2852. [PMID: 37232536 DOI: 10.1002/bit.28435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023]
Abstract
More than half of licensed therapeutic recombinant proteins (r-proteins) are manufactured using constitutively-expressing, stably-transfected Chinese hamster ovary (CHO) clones. While constitutive CHO expression systems have proven their efficacy for the manufacturing of monoclonal antibodies, many next-generation therapeutics such as cytokines and bispecific antibodies as well as biological targets such as ectodomains of transmembrane receptors remain intrinsically challenging to produce. Herein, we exploited a cumate-inducible CHO platform allowing reduced expression of various classes of r-proteins during selection of stable pools. Following stable pool generation, fed-batch productions showed that pools generated without cumate (OFF-pools) were significantly more productive than pools selected in the presence of cumate (ON-pools) for 8 out of the 10 r-proteins tested, including cytokines, G-protein coupled receptors (GPCRs), the HVEM membrane receptor ectodomain, the multifunctional protein High Mobility Group protein B1 (HMGB1), as well as monoclonal and bispecific T-cell engager antibodies. We showed that OFF-pools contain a significantly larger proportion of cells producing high levels of r-proteins and that these cells tend to proliferate faster when expression is turned off, suggesting that r-protein overexpression imposes a metabolic burden on the cells. Cell viability was lower and pool recovery was delayed during selection of ON-pools (mimicking constitutive expression), suggesting that high producers were likely lost or overgrown by faster-growing, low-producing cells. We also observed a correlation between the expression levels of the GPCRs with Binding immunoglobulin Protein, an endoplasmic reticulum (ER) stress marker. Taken together, these data suggest that using an inducible system to minimize r-protein expression during stable CHO pool selection reduces cellular stresses, including ER stress and metabolic burden, leading to pools with greater frequency of high-expressing cells, resulting in improved volumetric productivity.
Collapse
Affiliation(s)
- Jean-Sébastien Maltais
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Simon Lord-Dufour
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Audrey Morasse
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Matthew Stuible
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Martin Loignon
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Yves Durocher
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
17
|
Misorin AK, Chernyshova DO, Karbyshev MS. State-of-the-Art Approaches to Heterologous Expression of Bispecific Antibodies Targeting Solid Tumors. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1215-1231. [PMID: 37770390 DOI: 10.1134/s0006297923090031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/09/2023] [Accepted: 08/23/2023] [Indexed: 09/30/2023]
Abstract
Bispecific antibodies (bsAbs) are some of the most promising biotherapeutics due to the versatility provided by their structure and functional features. bsAbs simultaneously bind two antigens or two epitopes on the same antigen. Moreover, they are capable of directing immune effector cells to cancer cells and delivering various compounds (radionuclides, toxins, and immunologic agents) to the target cells, thus offering a broad spectrum of clinical applications. Current review is focused on the technologies used in bsAb engineering, current progress and prospects of these antibodies, and selection of various heterologous expression systems for bsAb production. We also discuss the platforms development of bsAbs for the therapy of solid tumors.
Collapse
|
18
|
Chi K, Xu H, Li H, Yang G, Zhou X, Gao XD. Expression of a Siglec-Fc Protein and Its Characterization. BIOLOGY 2023; 12:biology12040574. [PMID: 37106774 PMCID: PMC10135921 DOI: 10.3390/biology12040574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023]
Abstract
The emerging importance of the Siglec-sialic acid axis in human disease, especially cancer, has necessitated the identification of ligands for Siglecs. Recombinant Siglec-Fc fusion proteins have been widely used as ligand detectors, and also as sialic acid-targeted antibody-like proteins for cancer treatment. However, the heterogenetic properties of the Siglec-Fc fusion proteins prepared from various expression systems have not been fully elucidated. In this study, we selected HEK293 and CHO cells for producing Siglec9-Fc and further evaluated the properties of the products. The protein yield in CHO (8.23 mg/L) was slightly higher than that in HEK293 (7.46 mg/L). The Siglec9-Fc possesses five N-glycosylation sites and one of them is located in its Fc domain, which is important for the quality control of protein production and also the immunogenicity of Siglec-Fc. Our glycol-analysis confirmed that the recombinant protein from HEK293 received more fucosylation, while CHO showed more sialylation. Both products revealed a high dimerization ratio and sialic acid binding activity, which was confirmed by the staining of cancer cell lines and bladder cancer tissue. Finally, our Siglec9-Fc product was used to analyze the potential ligands on cancer cell lines.
Collapse
Affiliation(s)
- Kaijun Chi
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Huilin Xu
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Hanjie Li
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Ganglong Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoman Zhou
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Xiao-Dong Gao
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
19
|
Liang K, Luo H, Li Q. Enhancing and stabilizing monoclonal antibody production by Chinese hamster ovary (CHO) cells with optimized perfusion culture strategies. Front Bioeng Biotechnol 2023; 11:1112349. [PMID: 36741761 PMCID: PMC9895834 DOI: 10.3389/fbioe.2023.1112349] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/09/2023] [Indexed: 01/21/2023] Open
Abstract
The perfusion medium is critical in maintaining high cell concentration in cultures for the production of monoclonal antibody by Chinese hamster ovary cells. In this study, the effects of perfusion culture strategies when using different media on the process stability, product titer, and product quality were investigated in 3-L bioreactor. The results indicated that continuous perfusion could maintain higher levels of cell density, product titer, and quality in comparison with those of the intermittent perfusion culture. Next, the perfusion culture conditions with different perfusion rates and temperature reduction methods were further optimized. When combining the high perfusion rates and delayed reduction of culture temperature at day 6, the product titer reached a higher level of 16.19 g/L with the monomer relative abundant of 97.6%. In this case, the main peak of the product reached 56.3% and the total N-glycans ratio was 95.2%. To verify the effectiveness of the optimized perfusion culture in a larger scale, a 200-L bioreactor was used to perform and the final product titer reached the highest level of 16.79 g/L at day 16. Meanwhile, the product quality (monomer abundant of 97.6%, main peak of 56.3%, and N-glycans ratio of 96.5%) could also be well maintained. This study provided some guidance for the high-efficient production of monoclonal antibody by CHO cells via optimized perfusion culture strategy.
Collapse
Affiliation(s)
- Kexue Liang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Hongzhen Luo
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China,School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, China
| | - Qi Li
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China,*Correspondence: Qi Li,
| |
Collapse
|
20
|
Pirkalkhoran S, Grabowska WR, Kashkoli HH, Mirhassani R, Guiliano D, Dolphin C, Khalili H. Bioengineering of Antibody Fragments: Challenges and Opportunities. Bioengineering (Basel) 2023; 10:bioengineering10020122. [PMID: 36829616 PMCID: PMC9952581 DOI: 10.3390/bioengineering10020122] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Antibody fragments are used in the clinic as important therapeutic proteins for treatment of indications where better tissue penetration and less immunogenic molecules are needed. Several expression platforms have been employed for the production of these recombinant proteins, from which E. coli and CHO cell-based systems have emerged as the most promising hosts for higher expression. Because antibody fragments such as Fabs and scFvs are smaller than traditional antibody structures and do not require specific patterns of glycosylation decoration for therapeutic efficacy, it is possible to express them in systems with reduced post-translational modification capacity and high expression yield, for example, in plant and insect cell-based systems. In this review, we describe different bioengineering technologies along with their opportunities and difficulties to manufacture antibody fragments with consideration of stability, efficacy and safety for humans. There is still potential for a new production technology with a view of being simple, fast and cost-effective while maintaining the stability and efficacy of biotherapeutic fragments.
Collapse
Affiliation(s)
- Sama Pirkalkhoran
- School of Biomedical Science, University of West London, London W5 5RF, UK
| | | | | | | | - David Guiliano
- School of Life Science, College of Liberal Arts and Sciences, University of Westminster, London W1W 6UW, UK
| | - Colin Dolphin
- School of Biomedical Science, University of West London, London W5 5RF, UK
| | - Hanieh Khalili
- School of Biomedical Science, University of West London, London W5 5RF, UK
- School of Pharmacy, University College London, London WC1N 1AX, UK
- Correspondence:
| |
Collapse
|
21
|
Xu WJ, Lin Y, Mi CL, Pang JY, Wang TY. Progress in fed-batch culture for recombinant protein production in CHO cells. Appl Microbiol Biotechnol 2023; 107:1063-1075. [PMID: 36648523 PMCID: PMC9843118 DOI: 10.1007/s00253-022-12342-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 01/18/2023]
Abstract
Nearly 80% of the approved human therapeutic antibodies are produced by Chinese Hamster Ovary (CHO) cells. To achieve better cell growth and high-yield recombinant protein, fed-batch culture is typically used for recombinant protein production in CHO cells. According to the demand of nutrients consumption, feed medium containing multiple components in cell culture can affect the characteristics of cell growth and improve the yield and quality of recombinant protein. Fed-batch optimization should have a connection with comprehensive factors such as culture environmental parameters, feed composition, and feeding strategy. At present, process intensification (PI) is explored to maintain production flexible and meet forthcoming demands of biotherapeutics process. Here, CHO cell culture, feed composition in fed-batch culture, fed-batch culture environmental parameters, feeding strategies, metabolic byproducts in fed-batch culture, chemostat cultivation, and the intensified fed-batch are reviewed. KEY POINTS: • Fed-batch culture in CHO cells is reviewed. • Fed-batch has become a common technology for recombinant protein production. • Fed batch culture promotes recombinant protein production in CHO cells.
Collapse
Affiliation(s)
- Wen-Jing Xu
- grid.412990.70000 0004 1808 322XInternational Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003 Henan China ,grid.412990.70000 0004 1808 322XSchool of Pharmacy, Xinxiang Medical University, Xinxiang, 453003 Henan China
| | - Yan Lin
- grid.412990.70000 0004 1808 322XInternational Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003 Henan China ,grid.412990.70000 0004 1808 322XSchool of Nursing, Xinxiang Medical University, Xinxiang, 453003 Henan China
| | - Chun-Liu Mi
- grid.412990.70000 0004 1808 322XInternational Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003 Henan China
| | - Jing-Ying Pang
- grid.412990.70000 0004 1808 322XSchool of the First Clinical College, Xinxiang Medical University, Xinxiang, 453000 Henan China
| | - Tian-Yun Wang
- grid.412990.70000 0004 1808 322XInternational Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003 Henan China ,grid.495434.b0000 0004 1797 4346School of medicine, Xinxiang University, Xinxiang, 453003 Henan China
| |
Collapse
|
22
|
Kim K, Kim MG, Lee GM. Improving bone morphogenetic protein (BMP) production in CHO cells through understanding of BMP synthesis, signaling and endocytosis. Biotechnol Adv 2023; 62:108080. [PMID: 36526238 DOI: 10.1016/j.biotechadv.2022.108080] [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: 08/17/2022] [Revised: 12/01/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Bone morphogenetic proteins (BMPs) are a group of growth factors with the clinical potential to regulate cartilage and bone formation. Functionally active mature recombinant human BMPs (rhBMPs), produced primarily in Chinese hamster ovary (CHO) cells for clinical applications, are considered difficult to express because they undergo maturation processes, signaling pathways, or endocytosis. Although BMPs are a family of proteins with similar mature domain sequence identities, their individual properties are diverse. Thus, understanding the properties of individual rhBMPs is essential to improve rhBMP production in CHO cells. In this review, we discuss various approaches to improve rhBMP production in CHO cells by understanding the overall maturation process, signaling pathways and endocytosis of individual rhBMPs.
Collapse
Affiliation(s)
- Kyungsoo Kim
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Mi Gyeom Kim
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| |
Collapse
|
23
|
Stutz H. Advances and applications of electromigration methods in the analysis of therapeutic and diagnostic recombinant proteins – A Review. J Pharm Biomed Anal 2022; 222:115089. [DOI: 10.1016/j.jpba.2022.115089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022]
|
24
|
Richelle A, Corbett B, Agarwal P, Vernersson A, Trygg J, McCready C. Model-based intensification of CHO cell cultures: One-step strategy from fed-batch to perfusion. Front Bioeng Biotechnol 2022; 10:948905. [PMID: 36072286 PMCID: PMC9443430 DOI: 10.3389/fbioe.2022.948905] [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: 05/20/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
There is a growing interest in continuous processing of the biopharmaceutical industry. However, the technology transfer from traditional batch-based processes is considered a challenge as protocol and tools still remain to be established for their usage at the manufacturing scale. Here, we present a model-based approach to design optimized perfusion cultures of Chinese Hamster Ovary cells using only the knowledge captured during small-scale fed-batch experiments. The novelty of the proposed model lies in the simplicity of its structure. Thanks to the introduction of a new catch-all variable representing a bulk of by-products secreted by the cells during their cultivation, the model was able to successfully predict cellular behavior under different operating modes without changes in its formalism. To our knowledge, this is the first experimentally validated model capable, with a single set of parameters, to capture culture dynamic under different operating modes and at different scales.
Collapse
Affiliation(s)
- Anne Richelle
- Sartorius Corporate Research, Brussels, Belgium
- *Correspondence: Anne Richelle,
| | | | | | | | | | | |
Collapse
|
25
|
Kawabe Y, Kamihira M. Novel cell lines derived from Chinese hamster kidney tissue. PLoS One 2022; 17:e0266061. [PMID: 35358245 PMCID: PMC8970510 DOI: 10.1371/journal.pone.0266061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/13/2022] [Indexed: 01/15/2023] Open
Abstract
Immortalized kidney cell lines are widely used in basic and applied research such as cell permeability tests and drug screening. Although many cell lines have been established from kidney tissues, the immortalization process has not been clarified in these cell lines. In this study, we analyzed the phenotypic changes that occurred during the immortalization of kidney cells derived from Chinese hamster tissue in terms of karyotype and gene expression profiles. In the newly established cell line, designated as CHK-Q, gene expression profiles at each stage of the immortalization process and during the adaptation to serum-free conditions were analyzed by DNA microarray. Renal stem cell markers CD24 and CD133 were expressed in CHK-Q cells, suggesting that CHK-Q cells were transformed from renal stem cells. Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis to identify the pathways of upregulated and downregulated genes revealed that the immortalization of CHK-Q cells was associated with increased fluctuations in the expression of specific proto-oncogenes. Karyotype analysis of spontaneously immortalized CHK-Q cells indicated that CHK-Q chromosomes had a typical modal number of 23 but possessed slight chromosomal abnormalities. In this study, we investigated the mechanism of cell environmental adaptation by analyzing gene expression behavior during the immortalization process and serum-free adaptation. CHK-Q cells are applicable to the fields of biotechnology and biomedical science by utilizing their characteristics as kidney-derived cells.
Collapse
Affiliation(s)
- Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
- Manufacturing Technology Association of Biologics, Kobe, Japan
| | - Masamichi Kamihira
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
- Manufacturing Technology Association of Biologics, Kobe, Japan
- * E-mail:
| |
Collapse
|
26
|
Zhang JH, Shan LL, Liang F, Du CY, Li JJ. Strategies and Considerations for Improving Recombinant Antibody Production and Quality in Chinese Hamster Ovary Cells. Front Bioeng Biotechnol 2022; 10:856049. [PMID: 35316944 PMCID: PMC8934426 DOI: 10.3389/fbioe.2022.856049] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022] Open
Abstract
Recombinant antibodies are rapidly developing therapeutic agents; approximately 40 novel antibody molecules enter clinical trials each year, most of which are produced from Chinese hamster ovary (CHO) cells. However, one of the major bottlenecks restricting the development of antibody drugs is how to perform high-level expression and production of recombinant antibodies. The high-efficiency expression and quality of recombinant antibodies in CHO cells is determined by multiple factors. This review provides a comprehensive overview of several state-of-the-art approaches, such as optimization of gene sequence of antibody, construction and optimization of high-efficiency expression vector, using antibody expression system, transformation of host cell lines, and glycosylation modification. Finally, the authors discuss the potential of large-scale production of recombinant antibodies and development of culture processes for biopharmaceutical manufacturing in the future.
Collapse
Affiliation(s)
- Jun-He Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Recombinant Pharmaceutical Protein Expression System, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Jun-He Zhang,
| | - Lin-Lin Shan
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| | - Fan Liang
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
| | - Chen-Yang Du
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, China
| | - Jing-Jing Li
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
27
|
Physicochemical factors of bioprocessing impact the stability of therapeutic proteins. Biotechnol Adv 2022; 55:107909. [DOI: 10.1016/j.biotechadv.2022.107909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/09/2022] [Accepted: 01/09/2022] [Indexed: 02/06/2023]
|