1
|
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
|
2
|
Wang Q, Aliyu L, Chung CY, Rosenberg JN, Yu G, Betenbaugh MJ. Application of the CRISPR/Cas9 Gene Editing Method for Modulating Antibody Fucosylation in CHO Cells. Methods Mol Biol 2024; 2810:249-271. [PMID: 38926284 DOI: 10.1007/978-1-0716-3878-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Genetic engineering plays an essential role in the development of cell lines for biopharmaceutical manufacturing. Advanced gene editing tools can improve both the productivity of recombinant cell lines as well as the quality of therapeutic antibodies. Antibody glycosylation is a critical quality attribute for therapeutic biologics because the glycan patterns on the antibody fragment crystallizable (Fc) region can alter its clinical efficacy and safety as a therapeutic drug. As an example, recombinant antibodies derived from Chinese hamster ovary (CHO) cells are generally highly fucosylated; the absence of α1,6-fucose significantly enhances antibody-dependent cell-mediated cytotoxicity (ADCC) against cancer cells. This chapter describes a protocol applying clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) approach with different formats to disrupt the α-1,6-fucosyltransferase (FUT8) gene and subsequently inhibit α-1,6 fucosylation on antibodies expressed in CHO cells.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lateef Aliyu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Cheng-Yu Chung
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Julian N Rosenberg
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Geng Yu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
3
|
Gao Y, Ma Y, Xie D, Jiang H. ManNAc protects against podocyte pyroptosis via inhibiting mitochondrial damage and ROS/NLRP3 signaling pathway in diabetic kidney injury model. Int Immunopharmacol 2022; 107:108711. [PMID: 35338958 DOI: 10.1016/j.intimp.2022.108711] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/05/2022] [Accepted: 03/14/2022] [Indexed: 12/31/2022]
Abstract
Podocyte pyroptosis is an inflammatory form of cell death associated with Diabetic nephropathy (DN). It is reported that hyposialylated Angiopoietin-like-4 (Angptl4) secreted by glomerular podocytes plays an important role in the formation of proteinuria. Previous study indicated that supplementation of sialic acid precursor N-acetylmannosamine (ManNAc) could inhibit podocyte apoptosis and actin cytoskeleton rearrangement. Nevertheless, whether ManNAc could improve diabetic kidney damage by inhibiting podocyte pyroptosis remains unclear. This study aimed to explore the effect of ManNAc therapy on alleviating diabetic renal injury and podocyte pyroptosis, and its possible mechanism was also figured out. The male 8-week-old C57BL/6 mice were divided into three groups: control group, Streptozocin (STZ)-induced DN group, and ManNAc treated diabetic group. Then, the changes in renal function, renal histopathology, podocyte pyroptosis, reactive oxygen species (ROS), and mitochondrial dysfunction were measured. Herein, we observed that the upregulated expression of Angptl4 was involved in podocyte injury. ManNAc treatment ameliorated podocyte ultrastructure, renal function, and renal histopathology in STZ-induced DN mice. In addition, ManNAc administration attenuated podocyte cell death and suppressed the activation of Nucleotide leukin-rich polypeptide 3 (NLRP3), caspase-1, and interleukin-1β (IL-1β), and the cleavage of gasdermin-D (GSDMD). Moreover, ManNAc inhibited ROS production and restored mitochondrial morphology in vivo and vitro. Further, ManNAc administration significantly alleviated podocyte pyroptosis through inhibiting ROS/NLRP3 signaling pathway. Therefore, these results elucidated that the upregulated expression of Angptl4 was involved in podocyte injury and ManNAc treatment protected against podocyte pyroptosis via inhibiting mitochondrial injury and ROS/NLRP3 signaling pathway in DN mice.
Collapse
Affiliation(s)
- Yanmin Gao
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China; Department of General Practice, Kongjiang Community Health Service Center, Yangpu District, Shanghai 200093, China
| | - Yanli Ma
- Department of Pediatrics, Fourth People's Hospital Affiliated to Tongji University, Shanghai 200434, China
| | - Di Xie
- Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Hua Jiang
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| |
Collapse
|
4
|
Factors affecting the quality of therapeutic proteins in recombinant Chinese hamster ovary cell culture. Biotechnol Adv 2021; 54:107831. [PMID: 34480988 DOI: 10.1016/j.biotechadv.2021.107831] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/21/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022]
Abstract
Chinese hamster ovary (CHO) cells are the most widely used mammalian host cells for the commercial production of therapeutic proteins. Fed-batch culture is widely used to produce therapeutic proteins, including monoclonal antibodies, because of its operational simplicity and high product titer. Despite technical advances in the development of culture media and cell cultures, it is still challenging to maintain high productivity in fed-batch cultures while also ensuring good product quality. In this review, factors that affect the quality attributes of therapeutic proteins in recombinant CHO (rCHO) cell culture, such as glycosylation, charge variation, aggregation, and degradation, are summarized and categorized into three groups: culture environments, chemical additives, and host cell proteins accumulated in culture supernatants. Understanding the factors that influence the therapeutic protein quality in rCHO cell culture will facilitate the development of large-scale, high-yield fed-batch culture processes for the production of high-quality therapeutic proteins.
Collapse
|
5
|
Lemarié M, Chatonnet F, Caron G, Fest T. Early Emergence of Adaptive Mechanisms Sustaining Ig Production: Application to Antibody Therapy. Front Immunol 2021; 12:671998. [PMID: 33995412 PMCID: PMC8117215 DOI: 10.3389/fimmu.2021.671998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/12/2021] [Indexed: 01/13/2023] Open
Abstract
Antibody therapy, where artificially-produced immunoglobulins (Ig) are used to treat pathological conditions such as auto-immune diseases and cancers, is a very innovative and competitive field. Although substantial efforts have been made in recent years to obtain specific and efficient antibodies, there is still room for improvement especially when considering a precise tissular targeting or increasing antigen affinity. A better understanding of the cellular and molecular steps of terminal B cell differentiation, in which an antigen-activated B cell becomes an antibody secreting cell, may improve antibody therapy. In this review, we use our recently published data about human B cell differentiation, to show that the mechanisms necessary to adapt a metamorphosing B cell to its new secretory function appear quite early in the differentiation process i.e., at the pre-plasmablast stage. After characterizing the molecular pathways appearing at this stage, we will focus on recent findings about two main processes involved in antibody production: unfolded protein response (UPR) and endoplasmic reticulum (ER) stress. We’ll show that many genes coding for factors involved in UPR and ER stress are induced at the pre-plasmablast stage, sustaining our hypothesis. Finally, we propose to use this recently acquired knowledge to improve productivity of industrialized therapeutic antibodies.
Collapse
Affiliation(s)
- Maud Lemarié
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, Rennes, France
| | - Fabrice Chatonnet
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, Rennes, France.,Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, Rennes, France
| | - Gersende Caron
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, Rennes, France.,Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, Rennes, France
| | - Thierry Fest
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, Rennes, France.,Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, Rennes, France
| |
Collapse
|
6
|
Li W, Fan Z, Lin Y, Wang TY. Serum-Free Medium for Recombinant Protein Expression in Chinese Hamster Ovary Cells. Front Bioeng Biotechnol 2021; 9:646363. [PMID: 33791287 PMCID: PMC8006267 DOI: 10.3389/fbioe.2021.646363] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
At present, nearly 70% of recombinant therapeutic proteins (RTPs) are produced by Chinese hamster ovary (CHO) cells, and serum-free medium (SFM) is necessary for their culture to produce RTPs. In this review, the history and key components of SFM are first summarized, and its preparation and experimental design are described. Some small molecule compound additives can improve the yield and quality of RTP. The function and possible mechanisms of these additives are also reviewed here. Finally, the future perspectives of SFM use with CHO cells for RTP production are discussed.
Collapse
Affiliation(s)
- Weifeng Li
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| | - Zhenlin Fan
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yan Lin
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| | - Tian-Yun Wang
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China.,International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
7
|
Braasch K, Kryworuchko M, Piret JM. Autophagy-inducing peptide increases CHO cell monoclonal antibody production in batch and fed-batch cultures. Biotechnol Bioeng 2021; 118:1876-1883. [PMID: 33543765 DOI: 10.1002/bit.27703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 12/28/2022]
Abstract
The development of generic biopharmaceuticals is increasing the pressures for enhanced bioprocess productivity and yields. Autophagy ("self-eating") is a cellular process that allows cells to mitigate stresses such as nutrient deprivation. Reputed autophagy inhibitors have also been shown to increase autophagic flux under certain conditions, and enhance recombinant protein productivity in Chinese Hamster Ovary (CHO) cultures. Since peptides are commonly added to bioprocess culture media in hydrolysates, we evaluated the impact on productivity of an autophagy-inducing peptide (AIP), derived from the cellular autophagy protein Beclin 1. This was analyzed in CHO cell batch and fed-batch serum-free cultures producing a human Immunoglobulin G1 (IgG1). Interestingly, the addition of 1-4 µM AIP enhanced productivity in a concentration-dependent manner. Cell-specific productivity increased up to 1.8-fold in batch cultures, while in fed-batch cultures a maximum twofold increase in IgG concentration was observed. An initial drop in cell viability also occurred before cultures recovered normal growth. Overall, these findings strongly support the value of investigating the effects of autophagy pathway modulation, and in particular, the use of this AIP medium additive to increase CHO cell biotherapeutic protein production and yields.
Collapse
Affiliation(s)
- Katrin Braasch
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marko Kryworuchko
- BC Centre for Disease Control, Vancouver, British Columbia, Canada.,Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, British Columbia, Canada.,School of Public Health, Vaccinology and Immunotherapeutics, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - James M Piret
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
8
|
Violacein improves recombinant IgG production by controlling the cell cycle of Chinese hamster ovary cells. Cytotechnology 2020; 73:319-332. [PMID: 34149168 DOI: 10.1007/s10616-020-00434-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/28/2020] [Indexed: 10/22/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are used as host cells for industrial monoclonal antibody (mAb) production. Cell cycle control is an effective approach to increase mAb production in the cell culture. Violacein, a purple-colored pigment produced by microorganisms, has diverse bioactive properties and has been proposed for various industrial applications. In this study, we evaluated the potency of violacein for cell cycle control and improvement of recombinant immunoglobulin G (IgG) production in CHO cells. Compared with the control, 0.9 μM violacein in a 14-day fed-batch culture increased the maximum IgG concentration by 37.6% via increasing the specific production rate and cell longevity. Cell cycle analysis showed that violacein induced G1 and G2/M phase arrest. However, the G1 arrest was observed only on day 1, while G2/M arrest lasted more than 3 days, suggesting that G2/M arrest mediated the violacein-induced enhanced IgG production. Moreover, in line with the increased protein expression, the expression levels of IgG mRNA and nutrient metabolic rates were also increased. N-Linked glycosylation and charge variant profiles were barely affected by violacein treatment. Our results indicate that violacein affects the cell cycle of CHO cells and increases IgG production without changing product quality, showing promise as a mAb production enhancer in CHO cells. The study provides insight into violacein utilization in industrial mAb manufacturing and can help develop advanced, effective mAb production technologies using CHO cell cultures.
Collapse
|
9
|
Safari F, Farajnia S, Behzad Behbahani A, Zarredar H, Barekati-Mowahed M, Dehghani H. Caspase-7 deficiency in Chinese hamster ovary cells reduces cell proliferation and viability. Biol Res 2020; 53:52. [PMID: 33187557 PMCID: PMC7666471 DOI: 10.1186/s40659-020-00319-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/04/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Chinese hamster ovary (CHO) cells are the most commonly used mammalian host cell in the commercial-scale production of biopharmaceutical proteins. Modification of genes involved in apoptosis may improve the productivity of CHO cells. Executive caspases, including caspases 3 and 7, play critical roles in apoptosis. The effects of the ablation of the caspase 7 gene on proliferation and viability of CHO cells remains unknown. In this study, we applied clustered regularly interspaced short palindromic repeat (CRISPR/Cas9) to target caspase 7 gene of CHO K1 cell via all in one and homology targeted integration strategies. Consequently, the effect of caspase 7 deficiency on cell proliferation, viability, and apoptosis was studied by MTT assay and flow cytometry. RESULTS Findings of gel electrophoresis, western blotting, and sequencing confirmed the caspase 7 gene silencing in CHO cells (CHO-KO). Proliferation assay revealed that caspase 7 deficiency in CHO cells resulted in the reduction of proliferation in various CHO-KO clones. Besides, the disruption of caspase 7 had negative effects on cell viability in exposure with NaBu which confirmed by MTT assay. Results of flow cytometry using Anexin V/PI demonstrated that Nabu treatment (11 mM) declined the percentage of live CHO-K1 and CHO-KO cells to 70.3% and 5.79%. These results verified that the CHO-K1 cells were more resistant to apoptosis than CHO-KO, however most of CHO-KO cells undergone early apoptosis (91.9%) which seems to be a fascinating finding. CONCLUSION These results reveal that caspase 7 may be involved in the cell cycle progression of CHO cells. Furthermore, it seems that targeting caspase 7 is not the ideal route as it had previously been imagined within the prevention of apoptosis but the relation between caspase 7 deficiency, cell cycle arrest, and the occurrence of early apoptosis will require more investigation.
Collapse
Affiliation(s)
- Fatemeh Safari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Safar Farajnia
- Biotechnology Research Center, Tabriz University of Medical Sciences, Daneshgah Ave., Tabriz, Iran.
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Abbas Behzad Behbahani
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Habib Zarredar
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mazyar Barekati-Mowahed
- Department of Physiology & Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hesam Dehghani
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
10
|
Xu P, Xu S, He C, Khetan A. Applications of small molecules in modulating productivity and product quality of recombinant proteins produced using cell cultures. Biotechnol Adv 2020; 43:107577. [PMID: 32540474 DOI: 10.1016/j.biotechadv.2020.107577] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023]
Abstract
Mammalian cell cultures have been used extensively for production of recombinant protein therapeutics such as monoclonal antibodies, fusion proteins and enzymes for decades. Small molecules have been investigated as media supplements to improve process productivity and reduce cost of goods. Those chemicals can lead to significant yield improvement through different mechanisms such as cell cycle modulation, cellular redox regulation, etc. In addition to productivity, small molecules have also been routinely used to regulate post-translational modifications of recombinant proteins. This review summarizes key applications of small molecules in protein productivity improvement and product quality control.
Collapse
Affiliation(s)
- Ping Xu
- Biologics Development, Global Product Development & Supply, Bristol Myers Squibb Company, New Brunswick, NJ 08903, United States of America.
| | - Sen Xu
- Biologics Development, Global Product Development & Supply, Bristol Myers Squibb Company, New Brunswick, NJ 08903, United States of America
| | - Chunyan He
- Biologics Development, Global Product Development & Supply, Bristol Myers Squibb Company, New Brunswick, NJ 08903, United States of America
| | - Anurag Khetan
- Biologics Development, Global Product Development & Supply, Bristol Myers Squibb Company, New Brunswick, NJ 08903, United States of America
| |
Collapse
|
11
|
Xu J, Rehmann MS, Tian J, He Q, Chen J, Lee J, Borys MC, Li ZJ. Rosmarinic acid, a new raw material, doubled monoclonal antibody titer in cell culture manufacturing. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
12
|
Saeui CT, Cho KC, Dharmarha V, Nairn AV, Galizzi M, Shah SR, Gowda P, Park M, Austin M, Clarke A, Cai E, Buettner MJ, Ariss R, Moremen KW, Zhang H, Yarema KJ. Cell Line-, Protein-, and Sialoglycosite-Specific Control of Flux-Based Sialylation in Human Breast Cells: Implications for Cancer Progression. Front Chem 2020; 8:13. [PMID: 32117864 PMCID: PMC7013041 DOI: 10.3389/fchem.2020.00013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
Sialylation, a post-translational modification that impacts the structure, activity, and longevity of glycoproteins has been thought to be controlled primarily by the expression of sialyltransferases (STs). In this report we explore the complementary impact of metabolic flux on sialylation using a glycoengineering approach. Specifically, we treated three human breast cell lines (MCF10A, T-47D, and MDA-MB-231) with 1,3,4-O-Bu3ManNAc, a "high flux" metabolic precursor for the sialic acid biosynthetic pathway. We then analyzed N-glycan sialylation using solid phase extraction of glycopeptides (SPEG) mass spectrometry-based proteomics under conditions that selectively captured sialic acid-containing glycopeptides, referred to as "sialoglycosites." Gene ontology (GO) analysis showed that flux-based changes to sialylation were broadly distributed across classes of proteins in 1,3,4-O-Bu3ManNAc-treated cells. Only three categories of proteins, however, were "highly responsive" to flux (defined as two or more sialylation changes of 10-fold or greater). Two of these categories were cell signaling and cell adhesion, which reflect well-known roles of sialic acid in oncogenesis. A third category-protein folding chaperones-was unexpected because little precedent exists for the role of glycosylation in the activity of these proteins. The highly flux-responsive proteins were all linked to cancer but sometimes as tumor suppressors, other times as proto-oncogenes, or sometimes both depending on sialylation status. A notable aspect of our analysis of metabolically glycoengineered breast cells was decreased sialylation of a subset of glycosites, which was unexpected because of the increased intracellular levels of sialometabolite "building blocks" in the 1,3,4-O-Bu3ManNAc-treated cells. Sites of decreased sialylation were minor in the MCF10A (<25% of all glycosites) and T-47D (<15%) cells but dominated in the MDA-MB-231 line (~60%) suggesting that excess sialic acid could be detrimental in advanced cancer and cancer cells can evolve mechanisms to guard against hypersialylation. In summary, flux-driven changes to sialylation offer an intriguing and novel mechanism to switch between context-dependent pro- or anti-cancer activities of the several oncoproteins identified in this study. These findings illustrate how metabolic glycoengineering can uncover novel roles of sialic acid in oncogenesis.
Collapse
Affiliation(s)
- Christopher T Saeui
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kyung-Cho Cho
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Vrinda Dharmarha
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Alison V Nairn
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Melina Galizzi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Sagar R Shah
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Prateek Gowda
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Marian Park
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Melissa Austin
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Amelia Clarke
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Edward Cai
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Matthew J Buettner
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Ryan Ariss
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, United States
| | - Hui Zhang
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Kevin J Yarema
- Department of Biomedical Engineering, Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States.,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
13
|
Sharma S, Shekhar S, Sharma B, Jain P. Decoding glycans: deciphering the sugary secrets to be coherent on the implication. RSC Adv 2020; 10:34099-34113. [PMID: 35519023 PMCID: PMC9056758 DOI: 10.1039/d0ra04471g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/23/2020] [Indexed: 12/28/2022] Open
Abstract
Neoteric techniques, skills, and methodological advances in glycobiology and glycochemistry have been instrumental in pertinent discoveries to pave way for a new era in biomedical sciences. Glycans are sugar-based polymers that coat cells and decorate majority of proteins, forming glycoproteins. They are also found deposited in extracellular spaces between cells, attached to soluble signaling molecules, and are key players in several biological processes including regulation of immune responses and cell–cell interactions. Laboratory manipulations of protein, DNA and other macromolecules celebrate the accelerated research in respective fields, but the same seems unlikely for the complex sugar polymers. The structural complex polymers are neither synthesized using a known template nor are dynamically stable with respect to a cell's metabolic rate. What is more, sugar isomers—structurally distinct molecules with the same chemical formula—can be employed to construct varied glycans, but are almost impossible to tell apart based on molecular weight alone. The apparent lack of a glycan alphabet further reflects on an enduring question: how little do we know about the sugars? Evidently, glycan-based therapeutic potentials and glycomimetics are propitious advances for the future that have not been well exploited, and with a few conspicuous anomalies. Here, we contour the most notable contributions to enhance our ability to utilize the complex glycans as therapeutics. Diagnostic strategies concerning recurrent diseases and headways to address the challenges are also discussed. A glycan toolbox for pathogenic and cancerous interventions. The review article sheds light on the sweet secrets of this complex structure.![]()
Collapse
Affiliation(s)
- Shreya Sharma
- Department of Chemistry
- Netaji Subhas University of Technology
- India
| | - Shashank Shekhar
- Department of Chemistry
- Netaji Subhas University of Technology
- India
| | - Bhasha Sharma
- Department of Chemistry
- Netaji Subhas University of Technology
- India
| | - Purnima Jain
- Department of Chemistry
- Netaji Subhas University of Technology
- India
| |
Collapse
|
14
|
Dahodwala H, Kaushik P, Tejwani V, Kuo CC, Menard P, Henry M, Voldborg BG, Lewis NE, Meleady P, Sharfstein ST. Increased mAb production in amplified CHO cell lines is associated with increased interaction of CREB1 with transgene promoter. CURRENT RESEARCH IN BIOTECHNOLOGY 2019; 1:49-57. [PMID: 32577618 PMCID: PMC7311070 DOI: 10.1016/j.crbiot.2019.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Most therapeutic monoclonal antibodies in biopharmaceutical processes are produced in Chinese hamster ovary (CHO) cells. Technological advances have rendered the selection procedure for higher producers a robust protocol. However, information on molecular mechanisms that impart the property of hyper-productivity in the final selected clones is currently lacking. In this study, an IgG-producing industrial cell line and its methotrexate (MTX)-amplified progeny cell line were analyzed using transcriptomic, proteomic, phosphoproteomic, and chromatin immunoprecipitation (ChIP) techniques. Computational prediction of transcription factor binding to the transgene cytomegalovirus (CMV) promoter by the Transcription Element Search System and upstream regulator analysis of the differential transcriptomic data suggested increased in vivo CMV promoter-cAMP response element binding protein (CREB1) interaction in the higher producing cell line. Differential nuclear proteomic analysis detected 1.3-fold less CREB1 in the nucleus of the high productivity cell line compared with the parental cell line. However, the differential abundance of multiple CREB1 phosphopeptides suggested an increase in CREB1 activity in the higher producing cell line, which was confirmed by increased association of the CMV promotor with CREB1 in the high producer cell line. Thus, we show here that the nuclear proteome and phosphoproteome have an important role in regulating final productivity of recombinant proteins from CHO cells, and that CREB1 may play a role in transcriptional enhancement. Moreover, CREB1 phosphosites may be potential targets for cell engineering for increased productivity.
Collapse
Affiliation(s)
- Hussain Dahodwala
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY, USA
| | - Prashant Kaushik
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Vijay Tejwani
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY, USA
| | - Chih-Chung Kuo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Patrice Menard
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Bjorn G Voldborg
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Nathan E Lewis
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.,Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Susan T Sharfstein
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY, USA
| |
Collapse
|
15
|
Agatemor C, Buettner MJ, Ariss R, Muthiah K, Saeui CT, Yarema KJ. Exploiting metabolic glycoengineering to advance healthcare. Nat Rev Chem 2019; 3:605-620. [PMID: 31777760 DOI: 10.1038/s41570-019-0126-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Metabolic glycoengineering (MGE) is a technique for manipulating cellular metabolism to modulate glycosylation. MGE is used to increase the levels of natural glycans and, more importantly, to install non-natural monosaccharides into glycoconjugates. In this Review, we summarize the chemistry underlying MGE that has been developed over the past three decades and highlight several recent advances that have set the stage for clinical translation. In anticipation of near-term application to human healthcare, we describe emerging efforts to deploy MGE in diverse applications, ranging from the glycoengineering of biotherapeutic proteins and the diagnosis and treatment of complex diseases such as cancer to the development of new immunotherapies.
Collapse
Affiliation(s)
- Christian Agatemor
- Department of Biomedical Engineering and the Translational Tissue Engineering Center (TTEC), The Johns Hopkins University, Baltimore, MD, USA
| | - Matthew J Buettner
- Department of Biomedical Engineering and the Translational Tissue Engineering Center (TTEC), The Johns Hopkins University, Baltimore, MD, USA
| | - Ryan Ariss
- Department of Biomedical Engineering and the Translational Tissue Engineering Center (TTEC), The Johns Hopkins University, Baltimore, MD, USA
| | - Keerthana Muthiah
- Department of Biomedical Engineering and the Translational Tissue Engineering Center (TTEC), The Johns Hopkins University, Baltimore, MD, USA
| | - Christopher T Saeui
- Department of Biomedical Engineering and the Translational Tissue Engineering Center (TTEC), The Johns Hopkins University, Baltimore, MD, USA
| | - Kevin J Yarema
- Department of Biomedical Engineering and the Translational Tissue Engineering Center (TTEC), The Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
Wang Q, Yang G, Wang T, Yang W, Betenbaugh MJ, Zhang H. Characterization of intact glycopeptides reveals the impact of culture media on site-specific glycosylation of EPO-Fc fusion protein generated by CHO-GS cells. Biotechnol Bioeng 2019; 116:2303-2315. [PMID: 31062865 DOI: 10.1002/bit.27009] [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: 01/18/2019] [Revised: 04/15/2019] [Accepted: 05/02/2019] [Indexed: 01/08/2023]
Abstract
With the increasing demand to provide more detailed quality attributes, more sophisticated glycan analysis tools are highly desirable for biopharmaceutical manufacturing. Here, we performed an intact glycopeptide analysis method to simultaneously analyze the site-specific N- and O-glycan profiles of the recombinant erythropoietin Fc (EPO-Fc) protein secreted from a Chinese hamster ovary glutamine synthetase stable cell line and compared the effects of two commercial culture media, EX-CELL (EX) and immediate advantage (IA) media, on the glycosylation profile of the target protein. EPO-Fc, containing the Fc region of immunoglobulin G1 (IgG1) fused to EPO, was harvested at Day 5 and 8 of a batch cell culture process followed by purification and N- and O-glycopeptide profiling. A mixed anion exchange chromatographic column was implemented to capture and enrich N-linked glycopeptides. Using intact glycopeptide characterization, the EPO-Fc was observed to maintain their individual EPO and Fc N-glycan characteristics in which the EPO region presented bi-, tri-, and tetra-branched N-glycan structures, while the Fc N-glycan displayed mostly biantennary glycans. EPO-Fc protein generated in EX medium produced more complex tetra-antennary N-glycans at each of the three EPO N-sites while IA medium resulted in a greater fraction of bi- and tri-antennary N-glycans at these same sites. Interestingly, the sialylation content decreased from sites 1-4 in both media while the fucosylation progressively increased with a maximum at the final IgG Fc site. Moreover, we observed that low amounts of Neu5Gc were detected and the content increased at the later sampling time in both EX and IA media. For O-glycopeptides, both media produced predominantly three structures, N1F1F0SOG0, N1H1F0S1G0, and N1H1F0S2G0, with lesser amounts of other structures. This intact glycopeptide method can decipher site-specific glycosylation profile and provide a more detailed characterization of N- and O-glycans present for enhanced understanding of the key product quality attributes such as media on recombinant proteins of biotechnology interest.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Ganglong Yang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Tiexin Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Weiming Yang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
17
|
Sha S, Huang Z, Wang Z, Yoon S. Mechanistic modeling and applications for CHO cell culture development and production. Curr Opin Chem Eng 2018. [DOI: 10.1016/j.coche.2018.08.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
18
|
Buettner MJ, Shah SR, Saeui CT, Ariss R, Yarema KJ. Improving Immunotherapy Through Glycodesign. Front Immunol 2018; 9:2485. [PMID: 30450094 PMCID: PMC6224361 DOI: 10.3389/fimmu.2018.02485] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/08/2018] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy is revolutionizing health care, with the majority of high impact "drugs" approved in the past decade falling into this category of therapy. Despite considerable success, glycosylation-a key design parameter that ensures safety, optimizes biological response, and influences the pharmacokinetic properties of an immunotherapeutic-has slowed the development of this class of drugs in the past and remains challenging at present. This article describes how optimizing glycosylation through a variety of glycoengineering strategies provides enticing opportunities to not only avoid past pitfalls, but also to substantially improve immunotherapies including antibodies and recombinant proteins, and cell-based therapies. We cover design principles important for early stage pre-clinical development and also discuss how various glycoengineering strategies can augment the biomanufacturing process to ensure the overall effectiveness of immunotherapeutics.
Collapse
Affiliation(s)
- Matthew J Buettner
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Sagar R Shah
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Christopher T Saeui
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States.,Pharmacology/Toxicology Branch I, Division of Clinical Evaluation and Pharmacology/Toxicology, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Bethesda, MD, United States
| | - Ryan Ariss
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kevin J Yarema
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
19
|
Metabolic engineering of CHO cells to prepare glycoproteins. Emerg Top Life Sci 2018; 2:433-442. [DOI: 10.1042/etls20180056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 12/24/2022]
Abstract
As a complex and common post-translational modification, N-linked glycosylation affects a recombinant glycoprotein's biological activity and efficacy. For example, the α1,6-fucosylation significantly affects antibody-dependent cellular cytotoxicity and α2,6-sialylation is critical for antibody anti-inflammatory activity. Terminal sialylation is important for a glycoprotein's circulatory half-life. Chinese hamster ovary (CHO) cells are currently the predominant recombinant protein production platform, and, in this review, the characteristics of CHO glycosylation are summarized. Moreover, recent and current metabolic engineering strategies for tailoring glycoprotein fucosylation and sialylation in CHO cells, intensely investigated in the past decades, are described. One approach for reducing α1,6-fucosylation is through inhibiting fucosyltransferase (FUT8) expression by knockdown and knockout methods. Another approach to modulate fucosylation is through inhibition of multiple genes in the fucosylation biosynthesis pathway or through chemical inhibitors. To modulate antibody sialylation of the fragment crystallizable region, expressions of sialyltransferase and galactotransferase individually or together with amino acid mutations can affect antibody glycoforms and further influence antibody effector functions. The inhibition of sialidase expression and chemical supplementations are also effective and complementary approaches to improve the sialylation levels on recombinant glycoproteins. The engineering of CHO cells or protein sequence to control glycoforms to produce more homogenous glycans is an emerging topic. For modulating the glycosylation metabolic pathways, the interplay of multiple glyco-gene knockouts and knockins and the combination of multiple approaches, including genetic manipulation, protein engineering and chemical supplementation, are detailed in order to achieve specific glycan profiles on recombinant glycoproteins for superior biological function and effectiveness.
Collapse
|
20
|
Wang Q, Chung C, Yang W, Yang G, Chough S, Chen Y, Yin B, Bhattacharya R, Hu Y, Saeui CT, Yarema KJ, Betenbaugh MJ, Zhang H. Combining Butyrated ManNAc with Glycoengineered CHO Cells Improves EPO Glycan Quality and Production. Biotechnol J 2018; 14:e1800186. [DOI: 10.1002/biot.201800186] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 09/06/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Qiong Wang
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD 21218USA
| | - Cheng‐Yu Chung
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD 21218USA
| | - Weiming Yang
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMD 21231USA
| | - Ganglong Yang
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMD 21231USA
| | - Sandra Chough
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD 21218USA
| | - Yiqun Chen
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD 21218USA
| | - Bojiao Yin
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD 21218USA
| | - Rahul Bhattacharya
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMD 21231USA
| | - Yingwei Hu
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMD 21231USA
| | - Christopher T. Saeui
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMD 21231USA
| | - Kevin J. Yarema
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMD 21231USA
| | - Michael J. Betenbaugh
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD 21218USA
| | - Hui Zhang
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMD 21231USA
| |
Collapse
|
21
|
Wang Q, Chung CY, Rosenberg JN, Yu G, Betenbaugh MJ. Application of the CRISPR/Cas9 Gene Editing Method for Modulating Antibody Fucosylation in CHO Cells. Methods Mol Biol 2018; 1850:237-257. [PMID: 30242691 DOI: 10.1007/978-1-4939-8730-6_16] [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: 12/26/2022]
Abstract
Genetic engineering plays an essential role in the development of cell lines for biopharmaceutical manufacturing. Advanced gene editing tools can improve both the productivity of recombinant cell lines as well as the quality of therapeutic antibodies. Antibody glycosylation is a critical quality attribute for therapeutic biologics because the glycan patterns on the antibody fragment crystallizable (Fc) region can alter its clinical efficacy and safety as a therapeutic drug. As an example, recombinant antibodies derived from Chinese hamster ovary (CHO) cells are generally highly fucosylated; the absence of fucose significantly enhances antibody dependent cell-mediated cytotoxicity (ADCC) against cancer cells. This chapter describes a protocol applying clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) to disrupt the α-1,6-fucosyltranferase (FUT8) gene and subsequently inhibit α-1,6-fucosylation on antibodies expressed in CHO cells.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Cheng-Yu Chung
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Julian N Rosenberg
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Geng Yu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|