1
|
Sukwattananipaat P, Kuroda H, Yamano-Adachi N, Omasa T. Metabolomic characterization of monoclonal antibody-producing Chinese hamster lung (CHL)-YN cells in glucose-controlled serum-free fed-batch operation. Biotechnol Bioeng 2024; 121:2848-2867. [PMID: 39138873 DOI: 10.1002/bit.28777] [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: 03/08/2024] [Revised: 05/31/2024] [Accepted: 06/09/2024] [Indexed: 08/15/2024]
Abstract
The fast-growing Chinese hamster lung (CHL)-YN cell line was recently developed for monoclonal antibody production. In this study, we applied a serum-free fed-batch cultivation process to immunoglobulin (Ig)G1-producing CHL-YN cells, which were then used to design a dynamic glucose supply system to stabilize the extracellular glucose concentration based on glucose consumption. Glucose consumption of the cultures rapidly oscillated following three phases of glutamine metabolism: consumption, production, and re-consumption. Use of the dynamic glucose supply prolonged the viability of the CHL-YN-IgG1 cell cultures and increased IgG1 production. Liquid chromatography with tandem mass spectrometry-based target metabolomics analysis of the extracellular metabolites during the first glutamine shift was conducted to search for depleted compounds. The results suggest that the levels of four amino acids, namely arginine, aspartate, methionine, and serine, were sharply decreased in CHL-YN cells during glutamine production. Supporting evidence from metabolic and gene expression analyses also suggest that CHL-YN cells acquired ornithine- and cystathionine-production abilities that differed from those in Chinese hamster ovary-K1 cells, potentially leading to proline and cysteine biosynthesis.
Collapse
Affiliation(s)
| | - Hirotaka Kuroda
- Graduate School of Engineering, Osaka University, Osaka, Japan
- Shimadzu Corp., Kyoto, Japan
- Shimadzu Analytical Innovation Research Laboratories, Osaka, Japan
| | - Noriko Yamano-Adachi
- Graduate School of Engineering, Osaka University, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Manufacturing Technology Association of Biologics (MAB), Hyogo, Japan
| | - Takeshi Omasa
- Graduate School of Engineering, Osaka University, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Manufacturing Technology Association of Biologics (MAB), Hyogo, Japan
| |
Collapse
|
2
|
Toustou C, Boulogne I, Gonzalez AA, Bardor M. Comparative RNA-Seq of Ten Phaeodactylum tricornutum Accessions: Unravelling Criteria for Robust Strain Selection from a Bioproduction Point of View. Mar Drugs 2024; 22:353. [PMID: 39195469 DOI: 10.3390/md22080353] [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: 04/30/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
The production of biologics in mammalian cells is hindered by some limitations including high production costs, prompting the exploration of other alternative expression systems that are cheaper and sustainable like microalgae. Successful productions of biologics such as monoclonal antibodies have already been demonstrated in the diatom Phaeodactylum tricornutum; however, limited production yields still remain compared to mammalian cells. Therefore, efforts are needed to make this microalga more competitive as a cell biofactory. Among the seventeen reported accessions of P. tricornutum, ten have been mainly studied so far. Among them, some have already been used to produce high-value-added molecules such as biologics. The use of "omics" is increasingly being described as useful for the improvement of both upstream and downstream steps in bioprocesses using mammalian cells. Therefore, in this context, we performed an RNA-Seq analysis of the ten most used P. tricornutum accessions (Pt1 to Pt10) and deciphered the differential gene expression in pathways that could affect bioproduction of biologics in P. tricornutum. Our results highlighted the benefits of certain accessions such as Pt9 or Pt4 for the production of biologics. Indeed, these accessions seem to be more advantageous. Moreover, these results contribute to a better understanding of the molecular and cellular biology of P. tricornutum.
Collapse
Affiliation(s)
- Charlotte Toustou
- Laboratoire GlycoMEV UR 4358, Université de Rouen Normandie, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, 76000 Rouen, France
| | - Isabelle Boulogne
- Laboratoire GlycoMEV UR 4358, Université de Rouen Normandie, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, 76000 Rouen, France
| | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Muriel Bardor
- Laboratoire GlycoMEV UR 4358, Université de Rouen Normandie, SFR Normandie Végétal FED 4277, Innovation Chimie Carnot, 76000 Rouen, France
- ALGA BIOLOGICS, CURIB, 25 rue Tesnières, 76821 Mont-Saint-Aignan, France
| |
Collapse
|
3
|
Jerabek T, Weiß L, Fahrion H, Zeh N, Raab N, Lindner B, Fischer S, Otte K. In pursuit of a minimal CHO genome: Establishment of large-scale genome deletions. N Biotechnol 2024; 79:100-110. [PMID: 38154614 DOI: 10.1016/j.nbt.2023.12.007] [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: 10/20/2023] [Revised: 11/27/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
Chinese hamster ovary (CHO) cells are the most commonly used mammalian cell line for the production of complex therapeutic glycoproteins. As CHO cells have evolved as part of a multicellular organism, they harbor many cellular functions irrelevant for their application as production hosts in industrial bioprocesses. Consequently, CHO cells have been the target for numerous genetic engineering efforts in the past, but a tailored host cell chassis holistically optimized for its specific task in a bioreactor is still missing. While the concept of genome reduction has already been successfully applied to bacterial production cells, attempts to create higher eukaryotic production hosts exhibiting reduced genomes have not been reported yet. Here, we present the establishment and application of a large-scale genome deletion strategy for targeted excision of large genomic regions in CHO cells. We demonstrate the feasibility of genome reduction in CHO cells using optimized CRISPR/Cas9 based experimental protocols targeting large non-essential genomic regions with high efficiency. Achieved genome deletions of non-essential genetic regions did not introduce negative effects on bioprocess relevant parameters, although we conducted the largest reported genomic excision of 864 kilobase pairs in CHO cells so far. The concept presented serves as a directive to accelerate the development of a significantly genome-reduced CHO host cell chassis paving the way for a next generation of CHO cell factories.
Collapse
Affiliation(s)
- Tobias Jerabek
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany.
| | - Linus Weiß
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany
| | - Hannah Fahrion
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany
| | - Nikolas Zeh
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany; Boehringer Ingelheim Pharma GmbH & Co KG, Bioprocess Development Biologicals, Cell Line Development, Biberach, Germany
| | - Nadja Raab
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany
| | - Benjamin Lindner
- Boehringer Ingelheim Pharma GmbH & Co KG, Bioprocess Development Biologicals, Cell Line Development, Biberach, Germany
| | - Simon Fischer
- Boehringer Ingelheim Pharma GmbH & Co KG, Bioprocess Development Biologicals, Cell Line Development, Biberach, Germany
| | - Kerstin Otte
- University of Applied Sciences Biberach, Institute of Applied Biotechnology, Biberach, Germany
| |
Collapse
|
4
|
Liu HN, Wang XY, Zou Y, Wu WB, Lin Y, Ji BY, Wang TY. Synthetic enhancers including TFREs improve transgene expression in CHO cells. Heliyon 2024; 10:e26901. [PMID: 38468921 PMCID: PMC10926067 DOI: 10.1016/j.heliyon.2024.e26901] [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: 12/27/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/13/2024] Open
Abstract
The human cytomegalovirus major immediate early gene (CMV) promoter is currently the most preferred promoter for recombinant therapeutic proteins (RTPs) production in CHO cells. To enhance the production of RTPs, five synthetic enhancers including multiple transcription factor regulatory elements (TFREs) were evaluated to enhance recombinant protein level in transient and stably transfected CHO cells. Compared with the control, four elements can enhance the report genes expression under both two transfected states. Further, the function of these four enhancers on human serum albumin (HSA) were investigated. We found that the transient expression can increase by up to 1.5 times, and the stably expression can maximum increase by up to 2.14 times. The enhancement of transgene expression was caused by the boost of their corresponding mRNA levels. Transcriptomics analysis was performed and found that transcriptional activation and cell cycle regulation genes were involved. In conclusion, optimization of enhancers in the CMV promoter could increase the production yield of transgene in transfected CHO cells, which has significance for developing high-yield CHO cell expression system.
Collapse
Affiliation(s)
- Hui-Ning Liu
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
- SanQuan College of Xinxiang Medical University, Xinxiang 453003, China
| | - Xiao-Yin Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Ying Zou
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
| | - Wen-Bao Wu
- Shanghai Immunocan Biotech Co., LTD, Shanghai 200000, China
| | - Yan Lin
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
| | - Bo-Yu Ji
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
| | - Tian-Yun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang 453003, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
5
|
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
|
6
|
Luangwattananun P, Sangsuwannukul T, Supimon K, Thuwajit C, Chieochansin T, Sa-Nguanraksa D, Samarnthai N, O-Charoenrat P, Junking M, Yenchitsomanus PT. Anti-PD-L1 × anti-CD3 bispecific T-cell engager-armed T cells can overcome immunosuppression and redirect T cells to kill breast cancer cells expressing PD-L1. Int Immunopharmacol 2023; 124:111012. [PMID: 37804657 DOI: 10.1016/j.intimp.2023.111012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/09/2023]
Abstract
T cell-based immunotherapy has transformed cancer treatment. Nonetheless, T cell antitumor activity can be inhibited by an immune checkpoint molecule expressed on cancer cells, program death ligand 1 (PD-L1), which interacts with the PD-1 on T cells. We generated αPD-L1 × αCD3 bispecific T-cell engager-armed T cells (BATs) to prevent PD-L1/PD-1 interaction and hence to redirect T cells to kill cancer cells. αPD-L1 × αCD3 bispecific T-cell engagers (BTEs) were produced from Chinese hamster ovary (CHO) cells to arm human primary T cells. Flow cytometry was used to investigate BTE binding to BATs. The cytotoxicity of BATs against PD-L1-expressing breast cancer (BC) cell lines was assessed in 2-dimensional (2D) and 3-dimensional (3D) culture models. The binding stability of BTE on BATs and their efficacy after cryopreservation were also examined. The CHO cell BTE expression yield was 3.34 mg/ml. The binding ability on T cells reached 91.02 ± 4.2 %. BATs specifically lysed PD-L1-expressing BC cells, with 56.4 ± 15.3 % HCC70 cells and 70.67 ± 15.6 % MDA-MB-231 cells lysed at a 10:1 effector-to-target ratio. BATs showed slight, nonsignificant lysis of PD-L1-negative BC cells, MCF-7, and T47D. Moreover, BATs significantly disrupted MDA-MB-231 3D spheroids expressing PD-L1 after 48 and 72 h of coculture. Cryopreserved BATs maintained BTE binding stability, cell viability, and anticancer activity, comparable to fresh BATs. αPD-L1 × αCD3 BATs induced the cytolysis of PD-L1-expressing BC cells in 2D and 3D coculture assays. BATs can be prepared and preserved, facilitating their use and transportation. This study demonstrates the potential of αPD-L1 × αCD3 BATs in treating cancers with positive PD-L1 expression.
Collapse
Affiliation(s)
- Piriya Luangwattananun
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thanich Sangsuwannukul
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kamonlapat Supimon
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanitra Thuwajit
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thaweesak Chieochansin
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Doonyapat Sa-Nguanraksa
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Norasate Samarnthai
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Mutita Junking
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
7
|
Masson HO, Karottki KJLC, Tat J, Hefzi H, Lewis NE. From observational to actionable: rethinking omics in biologics production. Trends Biotechnol 2023; 41:1127-1138. [PMID: 37062598 PMCID: PMC10524802 DOI: 10.1016/j.tibtech.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 04/18/2023]
Abstract
As the era of omics continues to expand with increasing ubiquity and success in both academia and industry, omics-based experiments are becoming commonplace in industrial biotechnology, including efforts to develop novel solutions in bioprocess optimization and cell line development. Omic technologies provide particularly valuable 'observational' insights for discovery science, especially in academic research and industrial R&D; however, biomanufacturing requires a different paradigm to unlock 'actionable' insights from omics. Here, we argue the value of omic experiments in biotechnology can be maximized with deliberate selection of omic approaches and forethought about analysis techniques. We describe important considerations when designing and implementing omic-based experiments and discuss how systems biology analysis strategies can enhance efforts to obtain actionable insights in mammalian-based biologics production.
Collapse
Affiliation(s)
- Helen O Masson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | - Jasmine Tat
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Amgen Inc., Thousand Oaks, CA, USA
| | | | - Nathan E Lewis
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Fu Q, Polanco A, Lee YS, Yoon S. Critical challenges and advances in recombinant adeno-associated virus (rAAV) biomanufacturing. Biotechnol Bioeng 2023; 120:2601-2621. [PMID: 37126355 DOI: 10.1002/bit.28412] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/27/2023] [Accepted: 04/19/2023] [Indexed: 05/02/2023]
Abstract
Gene therapy is a promising therapeutic approach for genetic and acquired diseases nowadays. Among DNA delivery vectors, recombinant adeno-associated virus (rAAV) is one of the most effective and safest vectors used in commercial drugs and clinical trials. However, the current yield of rAAV biomanufacturing lags behind the necessary dosages for clinical and commercial use, which embodies a concentrated reflection of low productivity of rAAV from host cells, difficult scalability of the rAAV-producing bioprocess, and high levels of impurities materialized during production. Those issues directly impact the price of gene therapy medicine in the market, limiting most patients' access to gene therapy. In this context, the current practices and several critical challenges associated with rAAV gene therapy bioprocesses are reviewed, followed by a discussion of recent advances in rAAV-mediated gene therapy and other therapeutic biological fields that could improve biomanufacturing if these advances are integrated effectively into the current systems. This review aims to provide the current state-of-the-art technology and perspectives to enhance the productivity of rAAV while reducing impurities during production of rAAV.
Collapse
Affiliation(s)
- Qiang Fu
- Department of Biomedical Engineering and Biotechnology, The University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Ashli Polanco
- Department of Chemical Engineering, The University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Yong Suk Lee
- Department of Pharmaceutical Sciences, The University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Seongkyu Yoon
- Department of Chemical Engineering, The University of Massachusetts Lowell, Lowell, Massachusetts, USA
| |
Collapse
|
9
|
Novak N, Baumann M, Friss A, Cairns V, DeMaria C, Borth N. LncRNA analysis of mAb producing CHO clones reveals marker and engineering potential. Metab Eng 2023; 78:26-40. [PMID: 37196898 DOI: 10.1016/j.ymben.2023.05.003] [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: 04/27/2022] [Revised: 05/09/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Long non-coding RNAs (lncRNAs) are a potential new cell line engineering tool for improvement of yield and stability of CHO cells. In this study, we performed RNA sequencing of mAb producer CHO clones to study the lncRNA and protein coding transcriptome in relation to productivity. First, a robust linear model was used to identify genes correlating to productivity. To unravel specific patterns in expression of these genes, we employed weighted gene coexpression analysis (WGCNA) to find coexpressed modules, looking both for lncRNAs and coding genes. There was little overlap in the genes associated with productivity between the two products studied, possibly due to the difference in absolute range of productivity between the two mAbs. Therefore, we focused on the product with higher productivity and stronger candidate lncRNAs. To evaluate their potential as engineering targets, these candidate lncRNAs were transiently overexpressed or deleted by stable CRISPR Cas9 knock out both in a high and a low productivity subclone. We found that the thus achieved expression level of the identified lncRNAs, as confirmed by qPCR, does correlate well to productivity, so that they represent good markers that may be used for early clone selection. Additionally, we found that the deletion of one tested lncRNA region decreased viable cell density (VCD), prolonged culture time and increased cell size, final titer and specific productivity per cell. These results demonstrate the feasibility and usefulness of engineering lncRNA expression in production cell lines.
Collapse
Affiliation(s)
- Neža Novak
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; ACIB, Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Martina Baumann
- ACIB, Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Amy Friss
- Sanofi Biopharmaceutics Development, Framingham, MA, USA
| | - Victor Cairns
- Sanofi Biopharmaceutics Development, Framingham, MA, USA
| | | | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; ACIB, Austrian Centre of Industrial Biotechnology, Graz, Austria.
| |
Collapse
|
10
|
Cordova LT, Dahodwala H, Elliott KS, Baik J, Odenewelder DC, Nmagu D, Skelton BA, Uy L, Klaubert SR, Synoground BF, Chitwood DG, Dhara VG, Naik HM, Morris CS, Yoon S, Betenbaugh M, Coffman J, Swartzwelder F, Gillmeister MP, Harcum SW, Lee KH. Generation of reference cell lines, media, and a process platform for CHO cell biomanufacturing. Biotechnol Bioeng 2023; 120:715-725. [PMID: 36411514 DOI: 10.1002/bit.28290] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/03/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Due to the favorable attributes of Chinese hamster ovary (CHO) cells for therapeutic proteins and antibodies biomanufacturing, companies generate proprietary cells with desirable phenotypes. One key attribute is the ability to stably express multi-gram per liter titers in chemically defined media. Cell, media, and feed diversity has limited community efforts to translate knowledge. Moreover, academic, and nonprofit researchers generally cannot study "industrially relevant" CHO cells due to limited public availability, and the time and knowledge required to generate such cells. To address these issues, a university-industrial consortium (Advanced Mammalian Biomanufacturing Innovation Center, AMBIC) has acquired two CHO "reference cell lines" from different lineages that express monoclonal antibodies. These reference cell lines have relevant production titers, key performance outcomes confirmed by multiple laboratories, and a detailed technology transfer protocol. In commercial media, titers over 2 g/L are reached. Fed-batch cultivation data from shake flask and scaled-down bioreactors is presented. Using productivity as the primary attribute, two academic sites aligned with tight reproducibility at each site. Further, a chemically defined media formulation was developed and evaluated in parallel to the commercial media. The goal of this work is to provide a universal, industrially relevant CHO culture platform to accelerate biomanufacturing innovation.
Collapse
Affiliation(s)
- Lauren T Cordova
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Hussain Dahodwala
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA.,National Institute for Innovation in Manufacturing Biopharmaceuticals, Newark, Delaware, USA
| | - Kathryn S Elliott
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Jongyoun Baik
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA.,Department of Biological Sciences and Bioengineering, Inha University, Incheon, South Korea
| | | | - Douglas Nmagu
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Bradley A Skelton
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Lisa Uy
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Stephanie R Klaubert
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina, USA
| | | | - Dylan G Chitwood
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Venkata Gayatri Dhara
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Harnish Mukesh Naik
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Caitlin S Morris
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Michael Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | - Sarah W Harcum
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA.,National Institute for Innovation in Manufacturing Biopharmaceuticals, Newark, Delaware, USA
| |
Collapse
|
11
|
Lao N, Barron N. Enhancing recombinant protein and viral vector production in mammalian cells by targeting the YTHDF readers of N 6 -methyladenosine in mRNA. Biotechnol J 2023; 18:e2200451. [PMID: 36692010 DOI: 10.1002/biot.202200451] [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: 09/02/2022] [Revised: 12/20/2022] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
N6 -methyladenosine (m6A) is the most abundant internal modification on eukaryotic mRNA and has been implicated in a wide range of fundamental cellular processes. This modification is regulated and interpreted by a set of writer, eraser, and reader proteins. To date, there have been no reports on the potential of mRNA epigenetic regulators to influence recombinant protein expression in mammalian cells. In this study, the potential of manipulating the expression of the m6A YTH domain-containing readers, YTHDF1, 2 and 3 to improve recombinant protein yield based on their role in regulating mRNA stability and promoting translation were evaluated. Using siRNA-mediated gene depletion, cDNA over-expression, and methylation-specific RNA immunoprecipitation, it is demonstrated that (i) knock-down of YTHDF2 enhances (~2-fold) the levels of recombinant protein derived from GFP and EPO transgenes in CHO cells; (ii) the effects of YTHDF2 depletion on transgene expression is m6A-mediated; and (iii) YTHDF2 depletion, or over-expression of YTHDF1 increases viral protein expression and yield of infectious lentiviral (LV) particles (~2-3-fold) in HEK293 cells. We conclude that various transgenes can be subjected to regulation by m6A regulators in mammalian cell lines and that these findings demonstrate the utility of epitranscriptomic-based approaches to host cell line engineering for improved recombinant protein and viral vector production.
Collapse
Affiliation(s)
- Nga Lao
- National Institute for Bioprocessing Research and Training, Dublin, Ireland
| | - Niall Barron
- National Institute for Bioprocessing Research and Training, Dublin, Ireland.,School of Chemical and Bioprocess Engineering, University College Dublin, Dublin, Ireland
| |
Collapse
|
12
|
Kretzmer C, Narasimhan RL, Lal RD, Balassi V, Ravellette J, Kotekar Manjunath AK, Koshy JJ, Viano M, Torre S, Zanda VM, Kumravat M, Saldanha KMR, Chandranpillai H, Nihad I, Zhong F, Sun Y, Gustin J, Borgschulte T, Liu J, Razafsky D. De novo assembly and annotation of the CHOZN® GS -/- genome supports high-throughput genome-scale screening. Biotechnol Bioeng 2022; 119:3632-3646. [PMID: 36073082 PMCID: PMC9825924 DOI: 10.1002/bit.28226] [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: 03/16/2022] [Revised: 07/20/2022] [Accepted: 08/28/2022] [Indexed: 01/11/2023]
Abstract
Chinese hamster ovary (CHO) cells have been used as the industry standard for the production of therapeutic monoclonal antibodies for several decades. Despite significant improvements in commercial-scale production processes and media, the CHO cell has remained largely unchanged. Due to the cost and complexity of whole-genome sequencing and gene-editing it has been difficult to obtain the tools necessary to improve the CHO cell line. With the advent of next-generation sequencing and the discovery of the CRISPR/Cas9 system it has become more cost effective to sequence and manipulate the CHO genome. Here, we provide a comprehensive de novo assembly and annotation of the CHO-K1 based CHOZN® GS-/- genome. Using this platform, we designed, built, and confirmed the functionality of a whole genome CRISPR guide RNA library that will allow the bioprocessing community to design a more robust CHO cell line leading to the production of life saving medications in a more cost-effective manner.
Collapse
Affiliation(s)
- Corey Kretzmer
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| | - Rajagopalan Lakshmi Narasimhan
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Rahul Deva Lal
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Vincent Balassi
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| | - James Ravellette
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| | - Ajaya Kumar Kotekar Manjunath
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Jesvin Joy Koshy
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Marta Viano
- Istituto di Ricerche Biomediche “A. Marxer” RBM S.p.A.IvreaItaly
| | - Serena Torre
- Istituto di Ricerche Biomediche “A. Marxer” RBM S.p.A.IvreaItaly
| | - Valeria M. Zanda
- Istituto di Ricerche Biomediche “A. Marxer” RBM S.p.A.IvreaItaly
| | - Mausam Kumravat
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Keith Metelo Raul Saldanha
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Harikrishnan Chandranpillai
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Ifra Nihad
- Bioinformatics, IT R&D Applications, Merck (Sigma‐Aldrich Chemicals Pvt. Ltd., A subsidiary of Merck KGaA, Darmstadt, Germany)BangaloreIndia
| | - Fei Zhong
- Life Science Bioinformatics, IT, MilliporeSigmaSt. LouisMissouriUSA
| | - Yi Sun
- Bioinformatics, IT R&D Applications, MilliporeSigmaSt. LouisMissouriUSA
| | - Jason Gustin
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| | | | - Jiajian Liu
- Life Science Bioinformatics, IT, MilliporeSigmaSt. LouisMissouriUSA
| | - David Razafsky
- Upstream Research and Development, MilliporeSigmaSt. LouisMissouriUSA
| |
Collapse
|
13
|
Effects and mechanisms of animal-free hydrolysates on recombination protein yields in CHO cells. Appl Microbiol Biotechnol 2022; 106:7387-7396. [PMID: 36229612 DOI: 10.1007/s00253-022-12229-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/02/2022]
Abstract
Chinese hamster ovary (CHO) cells are the commonly used cell lines for producing recombinant therapeutic proteins (RTPs) because they possess post-translational modifications similar to human cells. Culture media are necessary for cell growth, and their quality affects the yields and quality of RTPs. Due to safety concerns for the complex purification of RTPs, the development of serum-free media (SFM) is necessary for CHO cells. To meet the need for CHO cells with higher cell density and RTP productivity with consistent product quality in large-scale suspension cultures, the optimization of SFM through adding some enzymatic animal-free hydrolysates (AFHs) is preferred. The AFHs can improve cell culture performance and product yield of RTPs without affecting their quality. Here, the effect and mechanism of various AFHs in improving CHO cell culture performance and protein expression are reviewed. KEY POINTS: • AFHs that improve the recombinant protein yield of CHO cells are reviewed. • AFHs improve recombinant protein yield via influencing cell performance. • The AFHs do not affect the quality of recombinant protein in CHO cells. • AFHs can provide nutrients, block cell cycle, and reduce oxidative stress.
Collapse
|
14
|
Bueno-Soler A, Palacios-Oliva J, Dorvignit-Pedroso D, Quintana-Cantillo A, Ramirez-Roque Y, Santo Tomas-Pompa J, Solazabal-Armstrong JA, Ruiz-Ramirez I, Mateo-de Acosta C, Boggiano-Ayo T, Lao-Gonzalez T. Production of an anti-TNFα antibody in murine myeloma cells by perfusion culture. Appl Microbiol Biotechnol 2022; 106:5007-5021. [PMID: 35835964 DOI: 10.1007/s00253-022-12052-4] [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: 10/14/2021] [Revised: 06/16/2022] [Accepted: 06/25/2022] [Indexed: 11/26/2022]
Abstract
Infliximab is a mouse/human chimeric IgG1 monoclonal antibody which recognizes the proinflammatory cytokine, tumor necrosis factor α (TNFα), and inhibits receptor interactions, thereby decreasing inflammation and autoimmune response in patients. This monoclonal antibody has been successfully used to treat rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis. However, the high treatment cost limits patient access to this biotherapy. One alternative to this problem is the use of biosimilars. In this work, we describe the stable expression and physicochemical characterization of an anti-TNFα antibody. While infliximab is produced in recombinant murine SP2/0 cells, our anti-TNFα IgG antibody was expressed in recombinant murine NS0 myeloma cells. The best anti-TNFα antibody-expressing clone was selected from three clone candidates based on the stability of IgG expression levels, specific productivity as well as TNFα-binding activity compared to commercial infliximab. Our results indicate that the selected cell clone, culture medium, and fermentation mode allowed for the production of an anti-TNFα antibody with similar characteristics to the reference commercially available product. An optimization of the selected culture medium by metabolomics may increase the volumetric productivity of the process to satisfy the demand for this product. Further experiments should be performed to evaluate the biological properties of this anti-TNFα antibody. KEY POINTS: • An anti-TNFα antibody was produced in NS0 cells using perfusion culture. • A proprietary chemically defined culture medium was used to replace commercially available protein-free medium. • The purified anti-TNFα antibody was comparable to the reference marketed product.
Collapse
Affiliation(s)
- Alexi Bueno-Soler
- Process Development Direction, Center of Molecular Immunology, 11600, Playa, Havana, Cuba
| | - Julio Palacios-Oliva
- Process Development Direction, Center of Molecular Immunology, 11600, Playa, Havana, Cuba
| | - Denise Dorvignit-Pedroso
- Immunobiology Direction, Center of Molecular Immunology, 11600, Playa, Havana, Cuba
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | | | - Yaima Ramirez-Roque
- Process Development Direction, Center of Molecular Immunology, 11600, Playa, Havana, Cuba
| | | | | | - Ingrid Ruiz-Ramirez
- Quality Control Direction, Center of Molecular Immunology, 11600, Playa, Havana, Cuba
| | - Cristina Mateo-de Acosta
- Immunobiology Direction, Center of Molecular Immunology, 11600, Playa, Havana, Cuba
- CIMAB S. A, 11600, Playa, Havana, Cuba
| | - Tammy Boggiano-Ayo
- Process Development Direction, Center of Molecular Immunology, 11600, Playa, Havana, Cuba
| | - Thailin Lao-Gonzalez
- Process Development Direction, Center of Molecular Immunology, 11600, Playa, Havana, Cuba.
- Animal Biotechnology Division, Center for Genetic Engineering and Biotechnology, 10600, Playa, Havana, Cuba.
| |
Collapse
|
15
|
Data-driven and model-guided systematic framework for media development in CHO cell culture. Metab Eng 2022; 73:114-123. [DOI: 10.1016/j.ymben.2022.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 11/21/2022]
|
16
|
Xu T, Zhang J, Wang T, Wang X. Recombinant antibodies aggregation and overcoming strategies in CHO cells. Appl Microbiol Biotechnol 2022; 106:3913-3922. [PMID: 35608667 DOI: 10.1007/s00253-022-11977-0] [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: 02/01/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/27/2022]
Abstract
Mammalian cell lines are frequently used as the preferred host cells for producing recombinant therapeutic proteins (RTPs) having post-translational modified modifications 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 (RTAs) are among the most important and promising RTPs for biomedical applications. A major limitation associated with the use of RTAs is their aggregation, which can be caused by a variety of factors; this results in a reduction of quality. RTA aggregations are especially concerning as they can trigger human immune responses in humans and may be fatal. Therefore, the mechanisms underlying RTA aggregation and measures for avoiding aggregation are interesting topics in RTAs research. In this review, we discuss recent progress in the field of RTAs aggregation, with a focus on factors that cause aggregation during RTA production and the development of strategies for overcoming RTA aggregation. KEY POINTS: • The recombinant antibody aggregation in mammalian cell systems is reviewed. • Intracellular environment and extracellular parameters influence recombinant antibody aggregation. • Reducing the aggregations can improve the quality of recombinant antibodies.
Collapse
Affiliation(s)
- Tingting Xu
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China.,The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, Henan, China
| | - Jihong 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
| | - Tianyun 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.
| | - Xiaoyin 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.
| |
Collapse
|
17
|
Dahodwala H, Amenyah SD, Nicoletti S, Henry M, Lees-Murdock DJ, Sharfstein ST. Evaluation of site-specific methylation of the CMV promoter and its role in CHO cell productivity of a recombinant monoclonal antibody. Antib Ther 2022; 5:121-129. [PMID: 35719211 PMCID: PMC9199181 DOI: 10.1093/abt/tbac010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/13/2022] [Accepted: 03/02/2022] [Indexed: 11/17/2022] Open
Abstract
We previously demonstrated that increased monoclonal antibody productivity in dihydrofolate reductase (DHFR)-amplified CHO cells correlates with phosphorylated transcription factor-cytomegalovirus (CMV) promoter interactions. In this article, we extend the characterization to include CMV promoter methylation and its influence on NFκB and CREB1 transcription factor binding to the CMV promoter in two families of DHFR-amplified CHO cell lines. CMV promoter methylation was determined using bisulfite sequencing. To overcome Sanger-sequencing limitations due to high CG bias and multiple transgenes copies, pyrosequencing was used to determine the frequency of methylated cytosines in regions proximal to and containing the NFκB and CREB1 transcription-factor consensus binding sites. Chromatin immunoprecipitation was performed to interrogate transcription factor–DNA interactions. Antibodies to CREB1 and NFκB were used to immunoprecipitate formaldehyde-crosslinked protein-DNA fractions, followed by reverse transcription quantitative real-time polymerase chain reaction to quantitate the number of copies of CMV-promoter DNA bound to the various transcription factors. The relative unmethylated fraction at the CREB1 and NFκB consensus binding sites determined by pyrosequencing was correlated with transcription factor binding as determined by chromatin immunoprecipitation. Azacytidine treatment reduced methylation in all treated samples, though not at all methylation sites, while increasing transcription. Distinct promoter methylation patterns arise upon clonal selection in different families of cell lines. In both cell line families, increased methylation was observed upon amplification. In one family, the NFκB binding-site methylation was accompanied by increased CREB1 interaction with the promoter. In the other cell line family, lower methylation frequency at the NFκB consensus binding site was accompanied by more NFκB recruitment to the promoter region.
Collapse
Affiliation(s)
- Hussain Dahodwala
- National Institute for Innovation in Manufacturing Biopharmaceuticals, Newark, Delaware, USA
| | - Sophia D Amenyah
- School of Biomedical Sciences, Ulster University, Coleraine, Londonderry, Northern Ireland, UK
| | - Sarah Nicoletti
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York USA
| | - Matthew Henry
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St. Lucia, QLD, Australia
| | - Diane J Lees-Murdock
- School of Biomedical Sciences, Ulster University, Coleraine, Londonderry, Northern Ireland, UK
| | - Susan T Sharfstein
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, New York USA
| |
Collapse
|
18
|
The potential of emerging sub-omics technologies for CHO cell engineering. Biotechnol Adv 2022; 59:107978. [DOI: 10.1016/j.biotechadv.2022.107978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/25/2022] [Accepted: 05/07/2022] [Indexed: 11/23/2022]
|
19
|
Kumar S, Kumar A, Huhn S, DeVine L, Cole R, Du Z, Betenbaugh M. A Proteomics Approach to Decipher a Sticky CHO Situation. Biotechnol Bioeng 2022; 119:2064-2075. [PMID: 35470426 PMCID: PMC9546176 DOI: 10.1002/bit.28108] [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: 11/18/2021] [Revised: 04/03/2022] [Accepted: 04/14/2022] [Indexed: 11/08/2022]
Abstract
Chinese hamster ovary (CHO) cells serve as protein therapeutics workhorses, so it is useful to understand what intrinsic properties make certain host cell lines and clones preferable for scale up and production of target proteins. In this study, two CHO host cell lines (H1, H2), and their respective clones were evaluated using comparative TMT‐proteomics. The clones obtained from host H1 showed increased productivity (6.8 times higher) in comparison to clones from host H2. Based on fold‐change analyses, we observed differential regulation in pathways including cell adhesion, aggregation, and cellular metabolism among others. In particular, the cellular adhesion pathway was downregulated in H1, in which podoplanin, an antiadhesion molecule, was upregulated the most in host H1 and associated clones. Phenotypically, these cells were less likely to aggregate and adhere to surfaces. In addition, enzymes involved in cellular metabolism such as isocitrate dehydrogenase (IDH) and mitochondrial‐d‐lactate dehydrogenase (
d‐LDHm) were also found to be differentially regulated. IDH plays a key role in TCA cycle and isocitrate‐alpha‐ketoglutarate cycle while
d‐LDHm aids in the elimination of toxic metabolite methylglyoxal, involved in protein degradation. These findings will enhance our efforts towards understanding why certain CHO cell lines exhibit enhanced performance and perhaps provide future cell engineering targets.
Collapse
Affiliation(s)
- Swetha Kumar
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Amit Kumar
- Process Cell Sciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Steven Huhn
- Process Cell Sciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Lauren DeVine
- Mass Spectrometry and Proteomics Facility, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Robert Cole
- Mass Spectrometry and Proteomics Facility, Johns Hopkins Medical Institute, Baltimore, MD, USA
| | - Zhimei Du
- Process Cell Sciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Michael Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
20
|
Spahn PN, Zhang X, Hu Q, Lu H, Hamaker NK, Hefzi H, Li S, Kuo CC, Huang Y, Lee JC, Davis AJ, Ly P, Lee KH, Lewis NE. Restoration of DNA repair mitigates genome instability and increases productivity of Chinese hamster ovary cells. Biotechnol Bioeng 2022; 119:963-982. [PMID: 34953085 PMCID: PMC8821244 DOI: 10.1002/bit.28016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/18/2021] [Indexed: 11/11/2022]
Abstract
Chinese hamster ovary (CHO) cells are the primary host for manufacturing of therapeutic proteins. However, productivity loss is a major problem and is associated with genome instability, as chromosomal aberrations reduce transgene copy number and decrease protein expression. We analyzed whole-genome sequencing data from 11 CHO cell lines and found deleterious single-nucleotide variants in DNA repair genes. Comparison with primary Chinese hamster cells confirmed DNA repair to be compromised in CHO. Correction of key DNA repair genes by single-nucleotide variant reversal or expression of intact complementary DNAs successfully improved DNA repair and mitigated karyotypic instability. Moreover, overexpression of intact copies of LIG4 and XRCC6 in a CHO cell line expressing secreted alkaline phosphatase mitigated transgene copy loss and improved protein titer retention. These results show that correction of DNA repair genes yields improvements in genome stability in CHO, and provide new opportunities for cell line development for sustainable protein expression.
Collapse
Affiliation(s)
- Philipp N. Spahn
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego School of Medicine, San Diego, La Jolla, CA 92093
| | - Xiaolin Zhang
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19711
| | - Qing Hu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Huiming Lu
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Nathaniel K. Hamaker
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19711
| | - Hooman Hefzi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Shangzhong Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Chih-Chung Kuo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Yingxiang Huang
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
| | - Jamie C. Lee
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Anthony J. Davis
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Peter Ly
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kelvin H. Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19711, These authors jointly supervised this work: Kelvin H. Lee, , 302-831-0344, Nathan E. Lewis, , 858-997-5844
| | - Nathan E. Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, The Novo Nordisk Foundation Center for Biosustainability at the University of California, San Diego School of Medicine, San Diego, La Jolla, CA 92093, Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, These authors jointly supervised this work: Kelvin H. Lee, , 302-831-0344, Nathan E. Lewis, , 858-997-5844
| |
Collapse
|
21
|
Marx N, Eisenhut P, Weinguny M, Klanert G, Borth N. How to train your cell - Towards controlling phenotypes by harnessing the epigenome of Chinese hamster ovary production cell lines. Biotechnol Adv 2022; 56:107924. [PMID: 35149147 DOI: 10.1016/j.biotechadv.2022.107924] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 11/24/2022]
Abstract
Recent advances in omics technologies and the broad availability of big datasets have revolutionized our understanding of Chinese hamster ovary cells in their role as the most prevalent host for production of complex biopharmaceuticals. In consequence, our perception of this "workhorse of the biopharmaceutical industry" has successively shifted from that of a nicely working, but unknown recombinant protein producing black box to a biological system governed by multiple complex regulatory layers that might possibly be harnessed and manipulated at will. Despite the tremendous progress that has been made to characterize CHO cells on various omics levels, our understanding is still far from complete. The well-known inherent genetic plasticity of any immortalized and rapidly dividing cell line also characterizes CHO cells and can lead to problematic instability of recombinant protein production. While the high mutational frequency has been a focus of CHO cell research for decades, the impact of epigenetics and its role in differential gene expression has only recently been addressed. In this review we provide an overview about the current understanding of epigenetic regulation in CHO cells and discuss its significance for shaping the cell's phenotype. We also look into current state-of-the-art technology that can be applied to harness and manipulate the epigenetic network so as to nudge CHO cells towards a specific phenotype. Here, we revise current strategies on site-directed integration and random as well as targeted epigenome modifications. Finally, we address open questions that need to be investigated to exploit the full repertoire of fine-tuned control of multiplexed gene expression using epigenetic and systems biology tools.
Collapse
Affiliation(s)
- Nicolas Marx
- University of Natural Resources and Life Sciences, Vienna, Austria
| | - Peter Eisenhut
- Austrian Centre for Industrial Biotechnology GmbH, Vienna, Austria
| | - Marcus Weinguny
- University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre for Industrial Biotechnology GmbH, Vienna, Austria
| | - Gerald Klanert
- Austrian Centre for Industrial Biotechnology GmbH, Vienna, Austria
| | - Nicole Borth
- University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Centre for Industrial Biotechnology GmbH, Vienna, Austria.
| |
Collapse
|
22
|
Verbelen B, Girardi T, Sulima SO, Vereecke S, Verstraete P, Verbeeck J, Royaert J, Cinque S, Montanaro L, Penzo M, Imbrechts M, Geukens N, Geuens T, Dierckx K, Pepe D, Kampen K, De Keersmaecker K. Exploitation of the ribosomal protein L10 R98S mutation to enhance recombinant protein production in mammalian cells. Eng Life Sci 2022; 22:100-114. [PMID: 35140557 PMCID: PMC8811726 DOI: 10.1002/elsc.202100124] [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: 09/20/2021] [Revised: 12/03/2021] [Accepted: 12/24/2021] [Indexed: 11/17/2022] Open
Abstract
Mammalian cells are commonly used to produce recombinant protein therapeutics, but suffer from a high cost per mg of protein produced. There is therefore great interest in improving protein yields to reduce production cost. We present an entirely novel approach to reach this goal through direct engineering of the cellular translation machinery by introducing the R98S point mutation in the catalytically essential ribosomal protein L10 (RPL10-R98S). Our data support that RPL10-R98S enhances translation levels and fidelity and reduces proteasomal activity in lymphoid Ba/F3 and Jurkat cell models. In HEK293T cells cultured in chemically defined medium, knock-in of RPL10-R98S was associated with a 1.7- to 2.5-fold increased production of four transiently expressed recombinant proteins and 1.7-fold for one out of two stably expressed proteins. In CHO-S cells, eGFP reached a 2-fold increased expression under stable but not transient conditions, but there was no production benefit for monoclonal antibodies. The RPL10-R98S associated production gain thus depends on culture conditions, cell type, and the nature of the expressed protein. Our study demonstrates the potential for using a ribosomal protein mutation for pharmaceutical protein production gains, and further research on how various factors influence RPL10-R98S phenotypes can maximize its exploitability for the mammalian protein production industry.
Collapse
Affiliation(s)
- Benno Verbelen
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Tiziana Girardi
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
- Flamingo TherapeuticsLeuvenBelgium
| | - Sergey O. Sulima
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
- Institute of Biological and Medical ImagingHelmholtz Zentrum München (GmbH)NeuherbergOberschleißheimGermany
- Center for Translational Cancer ResearchTechnical University of MunichMünchenGermany
| | - Stijn Vereecke
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Paulien Verstraete
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Jelle Verbeeck
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Jonathan Royaert
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Sonia Cinque
- Laboratory for RNA Cancer BiologyDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Lorenzo Montanaro
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department of ExperimentalDiagnostic and Specialty Medicine and Center for Applied Biomedical Research (CRBA)Alma Mater Studiorum‐University of BolognaBolognaItaly
| | - Marianna Penzo
- Department of ExperimentalDiagnostic and Specialty Medicine and Center for Applied Biomedical Research (CRBA)Alma Mater Studiorum‐University of BolognaBolognaItaly
| | - Maya Imbrechts
- Laboratory for Therapeutic and Diagnostic AntibodiesDepartment of Pharmaceutical and Pharmacological SciencesKU LeuvenLeuvenBelgium
| | - Nick Geukens
- Laboratory for Therapeutic and Diagnostic AntibodiesDepartment of Pharmaceutical and Pharmacological SciencesKU LeuvenLeuvenBelgium
- PharmAbsKU LeuvenLeuvenBelgium
| | | | | | - Daniele Pepe
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Kim Kampen
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
- Department of Radiotherapy, Maastricht Radiation Oncology (MAASTRO)Maastricht UniversityMaastrichtNetherlands
| | - Kim De Keersmaecker
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| |
Collapse
|
23
|
Tihanyi B, Nyitray L. Recent advances in CHO cell line development for recombinant protein production. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 38:25-34. [PMID: 34895638 DOI: 10.1016/j.ddtec.2021.02.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/02/2021] [Accepted: 02/23/2021] [Indexed: 12/20/2022]
Abstract
Recombinant proteins used in biomedical research, diagnostics and different therapies are mostly produced in Chinese hamster ovary cells in the pharmaceutical industry. These biotherapeutics, monoclonal antibodies in particular, have shown remarkable market growth in the past few decades. The increasing demand for high amounts of biologics requires continuous optimization and improvement of production technologies. Research aims at discovering better means and methods for reaching higher volumetric capacity, while maintaining stable product quality. An increasing number of complex novel protein therapeutics, such as viral antigens, vaccines, bi- and tri-specific monoclonal antibodies, are currently entering industrial production pipelines. These biomolecules are, in many cases, difficult to express and require tailored product-specific solutions to improve their transient or stable production. All these requirements boost the development of more efficient expression optimization systems and high-throughput screening platforms to facilitate the design of product-specific cell line engineering and production strategies. In this minireview, we provide an overview on recent advances in CHO cell line development, targeted genome manipulation techniques, selection systems and screening methods currently used in recombinant protein production.
Collapse
Affiliation(s)
- Borbála Tihanyi
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter stny 1/C, 1117 Budapest, Hungary
| | - László Nyitray
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter stny 1/C, 1117 Budapest, Hungary.
| |
Collapse
|
24
|
A Metabolomics Approach to Increasing Chinese Hamster Ovary (CHO) Cell Productivity. Metabolites 2021; 11:metabo11120823. [PMID: 34940581 PMCID: PMC8704136 DOI: 10.3390/metabo11120823] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Much progress has been made in improving the viable cell density of bioreactor cultures in monoclonal antibody production from Chinese hamster ovary (CHO) cells; however, specific productivity (qP) has not been increased to the same degree. In this work, we analyzed a library of 24 antibody-expressing CHO cell clones to identify metabolites that positively associate with qP and could be used for clone selection or medium supplementation. An initial library of 12 clones, each producing one of two antibodies, was analyzed using untargeted LC-MS experiments. Metabolic model-based annotation followed by correlation analysis detected 73 metabolites that significantly correlated with growth, qP, or both. Of these, metabolites in the alanine, aspartate, and glutamate metabolism pathway, and the TCA cycle showed the strongest association with qP. To evaluate whether these metabolites could be used as indicators to identify clones with potential for high productivity, we performed targeted LC-MS experiments on a second library of 12 clones expressing a third antibody. These experiments found that aspartate and cystine were positively correlated with qP, confirming the results from untargeted analysis. To investigate whether qP correlated metabolites reflected endogenous metabolic activity beneficial for productivity, several of these metabolites were tested as medium additives during cell culture. Medium supplementation with citrate improved qP by up to 490% and more than doubled the titer. Together, these studies demonstrate the potential for using metabolomics to discover novel metabolite additives that yield higher volumetric productivity in biologics production processes.
Collapse
|
25
|
MacDonald MA, Nöbel M, Roche Recinos D, Martínez VS, Schulz BL, Howard CB, Baker K, Shave E, Lee YY, Marcellin E, Mahler S, Nielsen LK, Munro T. Perfusion culture of Chinese Hamster Ovary cells for bioprocessing applications. Crit Rev Biotechnol 2021; 42:1099-1115. [PMID: 34844499 DOI: 10.1080/07388551.2021.1998821] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Much of the biopharmaceutical industry's success over the past 30 years has relied on products derived from Chinese Hamster Ovary (CHO) cell lines. During this time, improvements in mammalian cell cultures have come from cell line development and process optimization suited for large-scale fed-batch processes. Originally developed for high cell densities and sensitive products, perfusion processes have a long history. Driven by high volumetric titers and a small footprint, perfusion-based bioprocess research has regained an interest from academia and industry. The recent pandemic has further highlighted the need for such intensified biomanufacturing options. In this review, we outline the technical history of research in this field as it applies to biologics production in CHO cells. We demonstrate a number of emerging trends in the literature and corroborate these with underlying drivers in the commercial space. From these trends, we speculate that the future of perfusion bioprocesses is bright and that the fields of media optimization, continuous processing, and cell line engineering hold the greatest potential. Aligning in its continuous setup with the demands for Industry 4.0, perfusion biomanufacturing is likely to be a hot topic in the years to come.
Collapse
Affiliation(s)
- Michael A MacDonald
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia.,Thermo Fisher Scientific, Woolloongabba, Brisbane, Australia
| | - Matthias Nöbel
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia.,Thermo Fisher Scientific, Woolloongabba, Brisbane, Australia
| | - Dinora Roche Recinos
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia.,CSL Limited, Parkville, Melbourne, Australia
| | - Verónica S Martínez
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia
| | - Benjamin L Schulz
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Brisbane, Australia
| | - Christopher B Howard
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia
| | - Kym Baker
- Thermo Fisher Scientific, Woolloongabba, Brisbane, Australia
| | - Evan Shave
- Thermo Fisher Scientific, Woolloongabba, Brisbane, Australia
| | | | - Esteban Marcellin
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia.,Metabolomics Australia, The University of Queensland, St. Lucia, Brisbane, Australia
| | - Stephen Mahler
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia
| | - Lars Keld Nielsen
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia.,Metabolomics Australia, The University of Queensland, St. Lucia, Brisbane, Australia.,The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Trent Munro
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Australia.,National Biologics Facility, The University of Queensland, St. Lucia, Brisbane, Australia
| |
Collapse
|
26
|
Proteomic Landscape of Adeno-Associated Virus (AAV)-Producing HEK293 Cells. Int J Mol Sci 2021; 22:ijms222111499. [PMID: 34768929 PMCID: PMC8584267 DOI: 10.3390/ijms222111499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated viral (AAV) vectors are widely used for gene therapy, providing treatment for diseases caused by absent or defective genes. Despite the success of gene therapy, AAV manufacturing is still challenging, with production yields being limited. With increased patient demand, improvements in host cell productivity through various engineering strategies will be necessary. Here, we study the host cell proteome of AAV5-producing HEK293 cells using reversed phase nano-liquid chromatography and tandem mass spectrometry (RPLC-MS/MS). Relative label-free quantitation (LFQ) was performed, allowing a comparison of transfected vs. untransfected cells. Gene ontology enrichment and pathway analysis revealed differential expression of proteins involved in fundamental cellular processes such as metabolism, proliferation, and cell death. Furthermore, changes in expression of proteins involved in endocytosis and lysosomal degradation were observed. Our data provides highly valuable insights into cellular mechanisms involved during recombinant AAV production by HEK293 cells, thus potentially enabling further improvements of gene therapy product manufacturing.
Collapse
|
27
|
Rish AJ, Drennen JK, Anderson CA. Metabolic trends of Chinese hamster ovary cells in biopharmaceutical production under batch and fed-batch conditions. Biotechnol Prog 2021; 38:e3220. [PMID: 34676699 DOI: 10.1002/btpr.3220] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/05/2021] [Accepted: 10/19/2021] [Indexed: 11/07/2022]
Abstract
Extensive knowledge of Chinese hamster ovary (CHO) cell metabolism is required to improve process productivity and culture performance in biopharmaceutical manufacturing. However, CHO cells show a dynamic metabolism during culturing in batch and fed-batch bioreactors. CHO cell metabolism is generally described as taking place in three stages: exponential growth phase, stationary phase, and death phase. This review aims to summarize the trends of central metabolism for CHO cells during each stage. Additional insights into how culture conditions are related to phase transitions and force metabolic rewiring are provided. Understanding of CHO cell metabolism lends itself to improving culture qualities by, for example, identifying sources of toxic byproducts and pathways for cellular engineering. In summary, this review describes the changes in CHO cell central metabolism over the course of the culture.
Collapse
Affiliation(s)
- Adam J Rish
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, Pennsylvania, USA
| | - James K Drennen
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, Pennsylvania, USA
- Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Carl A Anderson
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, Pennsylvania, USA
- Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
28
|
Tao W, Ahmed W, Guo M, Mohsin A, Wu B, Li R. Selection of high-producing clones by a relative titer predictive model using image analysis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1144. [PMID: 34430585 PMCID: PMC8350677 DOI: 10.21037/atm-21-2822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/28/2021] [Indexed: 11/24/2022]
Abstract
Background The commercial success of monoclonal antibodies (Mabs) has made biological therapeutics attractive to pharmaceutical companies. The priority of biopharmaceutical companies is to acquire and develop cell lines that enable them to manufacture biologics quickly, consistently, and economically. Clone selection is a critical process for cell line development. However, the traditional clone selection process requires the evaluation of large numbers of clones using cell growth rate, cell densities and titer, product quality, and so on. Methods To improve efficiency of the clone selection strategies, we developed a relative titer (RT) prediction model by the quantitative information extracted from microscope images during the cell line development process. The performance of this RT prediction model was further evaluated with 50 clones from 5 different cell lines. Results The RT prediction model was able to predict high producers from a given data set when the same host cells were used. Although inaccurate prediction occurred when different host cell was used, this RT prediction model may serve as an excellent proof of concept study that quantitative information from cell line development images provides valuable information to facilitate the cell line development process. Conclusions Here, we present the first predictive model that can be used to estimate the relative productivity of Chinese hamster ovaries (CHO) clones during the cell line development. Additional experiments are currently in process to further improve the RT predictive model. Nevertheless, our current study will serve as a foundation for more prediction models for cell line development that can facilitate the selection of clones.
Collapse
Affiliation(s)
- Weihong Tao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Waqas Ahmed
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Bing Wu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Rongxiu Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
29
|
Schmieder V, Novak N, Dhiman H, Nguyen LN, Serafimova E, Klanert G, Baumann M, Kildegaard HF, Borth N. A pooled CRISPR/AsCpf1 screen using paired gRNAs to induce genomic deletions in Chinese hamster ovary cells. ACTA ACUST UNITED AC 2021; 31:e00649. [PMID: 34277363 PMCID: PMC8261548 DOI: 10.1016/j.btre.2021.e00649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/06/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022]
Abstract
• Development of a small-scale CRISPR/AsCpf1 screen in CHO. • Usage of paired gRNAs enables full deletion of coding or noncoding genomic regions. • Growth perturbing paired gRNAs identified. • Key points for considerations in future screens identified.
Chinese hamster ovary (CHO) cells are the most widely used host for the expression of therapeutic proteins. Recently, significant progress has been made due to advances in genome sequence and annotation quality to unravel the black box CHO. Nevertheless, in many cases the link between genotype and phenotype in the context of suspension cultivated production cell lines is still not fully understood. While frameshift approaches targeting coding genes are frequently used, the non-coding regions of the genome have received less attention with respect to such functional annotation. Importantly, for non-coding regions frameshift knock-out strategies are not feasible. In this study, we developed a CRISPR-mediated screening approach that performs full deletions of genomic regions to enable the functional study of both the translated and untranslated genome. An in silico pipeline for the computational high-throughput design of paired guide RNAs (pgRNAs) directing CRISPR/AsCpf1 was established and used to generate a library tackling process-related genes and long non-coding RNAs. Next generation sequencing analysis of the plasmid library revealed a sufficient, but highly variable pgRNA composition. Recombinase-mediated cassette exchange was applied for pgRNA library integration rather than viral transduction to ensure single copy representation of pgRNAs per cell. After transient AsCpf1 expression, cells were cultivated over two sequential batches to identify pgRNAs which massively affected growth and survival. By comparing pgRNA abundance, depleted candidates were identified and individually validated to verify their effect.
Collapse
Key Words
- AsCpf1, Cpf1 from Acidaminococcus sp BV3L6
- CHO, Chinese hamster ovary
- CPM, counts per million reads mapped
- CRISPR, Clustered Regularly Interspaced Short Palindromic Repeats
- CRISPR/AsCpf1
- Cas9, CRISPR-associated protein 9
- Chinese hamster ovary cells
- Cpf1, CRISPR-associated protein in Prevotella and Francisella
- DE, differentially expressed
- DOWN-TTS, downstream transcription termination site
- DR, differentially represented
- EV, empty vector
- EpoFc, Erythropoietin Fc fusion protein
- FACS, fluorescence activated cell sorting
- FC, fold change
- FDR, false discovery rate
- GS, glutamine synthetase
- Genetic screen
- NGS, next generation sequencing
- NTC, no template control
- PAM, protospacer adjacent motif
- PCA, principal component analysis
- Qp, specific productivity
- RMCE, recombinase-mediated cassette exchange
- TMM, trimmed mean of M values
- UP-TSS, upstream transcription start site
- VCD, viable cell density
- dCas9, deactivated Cas9
- gRNA, guide RNA
- genomic deletion
- lncRNA, long non-coding RNA
- ncGene, non-coding gene
- oligo, oligonucleotide
- paired gRNAs
- pgRNA, paired gRNA
- sgRNA, single guide RNA
- µ, growth rate
Collapse
Affiliation(s)
- Valerie Schmieder
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria.,acib GmbH, Austrian Centre of Industrial Biotechnology, Muthgasse 11, Vienna, Austria
| | - Neža Novak
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria.,acib GmbH, Austrian Centre of Industrial Biotechnology, Muthgasse 11, Vienna, Austria
| | - Heena Dhiman
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria.,acib GmbH, Austrian Centre of Industrial Biotechnology, Muthgasse 11, Vienna, Austria
| | - Ly Ngoc Nguyen
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria.,acib GmbH, Austrian Centre of Industrial Biotechnology, Muthgasse 11, Vienna, Austria
| | - Evgenija Serafimova
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria.,acib GmbH, Austrian Centre of Industrial Biotechnology, Muthgasse 11, Vienna, Austria
| | - Gerald Klanert
- acib GmbH, Austrian Centre of Industrial Biotechnology, Muthgasse 11, Vienna, Austria
| | - Martina Baumann
- acib GmbH, Austrian Centre of Industrial Biotechnology, Muthgasse 11, Vienna, Austria
| | - Helene Faustrup Kildegaard
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, Kgs. Lyngby, Denmark
| | - Nicole Borth
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria.,acib GmbH, Austrian Centre of Industrial Biotechnology, Muthgasse 11, Vienna, Austria
| |
Collapse
|
30
|
Pérez-Rodriguez S, Ramírez-Lira MDJ, Trujillo-Roldán MA, Valdez-Cruz NA. Nutrient supplementation strategy improves cell concentration and longevity, monoclonal antibody production and lactate metabolism of Chinese hamster ovary cells. Bioengineered 2021; 11:463-471. [PMID: 32223359 PMCID: PMC7161567 DOI: 10.1080/21655979.2020.1744266] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A careful selection of culture mediums and feeds has become necessary to maximize yields of recombinant proteins during bioprocesses of mammalian cells. Supplements contain a variety of concentrate nutrients, and their beneficial effects vary according to recombinant cell lines. In this study, the effects of PowerFeed A on growth kinetics, productivity and cellular metabolism were evaluated for two Chinese hamster ovary cell lines producing a monoclonal antibody in a batch culture. Supplemented cultures increased integral viable cell density of CRL-12444 and CRL-12445 cells by 2.4 and 1.6 times through extension of culture time at which viability was above 90% in 72 and 36 h, respectively, and increment of maximal cell concentration in 3.25 × 106 cells/ml (69%) for CRL-12445 cells. Product titer augmented 1.9 and 2.5 times for CRL-12444 and CRL-12445 cells, respectively, without changes in growth rate and specific productivity. Feed supplementation also stimulated full consumption of glucose and free glutamine and reduced 10 times lactate accumulation, while ammonium, sodium and potassium remained at similar concentrations at the end of the culture. About 44% of calcium, mainly provided by feed, was consumed by both cell lines. Maximization of cellular growth, viability and protein titer through feeding encourages extending its use to other cell lines and exploring novel combinations with other basal mediums or feeds. A thorough investigation of its impact on protein quality and the molecular mechanisms behind these effects will allow designing effective feeds and strategies to rationally optimize protein production in the biomanufacturing industry.
Collapse
Affiliation(s)
- Saumel Pérez-Rodriguez
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, Ciudad De México, México
| | - María de Jesús Ramírez-Lira
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, Ciudad De México, México
| | - Mauricio A Trujillo-Roldán
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, Ciudad De México, México
| | - Norma A Valdez-Cruz
- Programa de Investigación de Producción de Biomoléculas, Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, Ciudad De México, México
| |
Collapse
|
31
|
Donini R, Haslam SM, Kontoravdi C. Glycoengineering Chinese hamster ovary cells: a short history. Biochem Soc Trans 2021; 49:915-931. [PMID: 33704400 PMCID: PMC8106501 DOI: 10.1042/bst20200840] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/26/2021] [Accepted: 02/08/2021] [Indexed: 12/25/2022]
Abstract
Biotherapeutic glycoproteins have revolutionised the field of pharmaceuticals, with new discoveries and continuous improvements underpinning the rapid growth of this industry. N-glycosylation is a critical quality attribute of biotherapeutic glycoproteins that influences the efficacy, half-life and immunogenicity of these drugs. This review will focus on the advances and future directions of remodelling N-glycosylation in Chinese hamster ovary (CHO) cells, which are the workhorse of recombinant biotherapeutic production, with particular emphasis on antibody products, using strategies such as cell line and protein backbone engineering.
Collapse
Affiliation(s)
- Roberto Donini
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| | - Stuart M. Haslam
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
| | - Cleo Kontoravdi
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| |
Collapse
|
32
|
Bryan L, Clynes M, Meleady P. The emerging role of cellular post-translational modifications in modulating growth and productivity of recombinant Chinese hamster ovary cells. Biotechnol Adv 2021; 49:107757. [PMID: 33895332 DOI: 10.1016/j.biotechadv.2021.107757] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023]
Abstract
Chinese hamster ovary (CHO) cells are one of the most commonly used host cell lines used for the production human therapeutic proteins. Much research over the past two decades has focussed on improving the growth, titre and cell specific productivity of CHO cells and in turn lowering the costs associated with production of recombinant proteins. CHO cell engineering has become of particular interest in recent years following the publication of the CHO cell genome and the availability of data relating to the proteome, transcriptome and metabolome of CHO cells. However, data relating to the cellular post-translational modification (PTMs) which can affect the functionality of CHO cellular proteins has only begun to be presented in recent years. PTMs are important to many cellular processes and can further alter proteins by increasing the complexity of proteins and their interactions. In this review, we describe the research presented from CHO cells to date related on three of the most important PTMs; glycosylation, phosphorylation and ubiquitination.
Collapse
Affiliation(s)
- Laura Bryan
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
33
|
Strasser L, Farrell A, Ho JTC, Scheffler K, Cook K, Pankert P, Mowlds P, Viner R, Karger BL, Bones J. Proteomic Profiling of IgG1 Producing CHO Cells Using LC/LC-SPS-MS 3: The Effects of Bioprocessing Conditions on Productivity and Product Quality. Front Bioeng Biotechnol 2021; 9:569045. [PMID: 33898396 PMCID: PMC8062983 DOI: 10.3389/fbioe.2021.569045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
The biopharmaceutical market is dominated by monoclonal antibodies, the majority of which are produced in Chinese hamster ovary (CHO) cell lines. Intense cell engineering, in combination with optimization of various process parameters results in increasing product titers. To enable further improvements in manufacturing processes, detailed information about how certain parameters affect cellular mechanisms in the production cells, and thereby also the expressed drug substance, is required. Therefore, in this study the effects of commonly applied changes in bioprocessing parameters on an anti-IL8 IgG1 producing CHO DP-12 cell line were investigated on the level of host cell proteome expression combined with product quality assessment of the expressed IgG1 monoclonal antibody. Applying shifts in temperature, pH and dissolved oxygen concentration, respectively, resulted in altered productivity and product quality. Furthermore, analysis of the cells using two-dimensional liquid chromatography-mass spectrometry employing tandem mass tag based isotopic quantitation and synchronous precursor selection-MS3 detection revealed substantial changes in the protein expression profiles of CHO cells. Pathway analysis indicated that applied bioprocessing conditions resulted in differential activation of oxidative phosphorylation. Additionally, activation of ERK5 and TNFR1 signaling suggested an affected cell cycle. Moreover, in-depth product characterization by means of charge variant analysis, peptide mapping, as well as structural and functional analysis, revealed posttranslational and structural changes in the expressed drug substance. Taken together, the present study allows the conclusion that, in anti-IL8 IgG1 producing CHO DP-12 cells, an improved energy metabolism achieved by lowering the cell culture pH is favorable when aiming towards high antibody production rates while maintaining product quality.
Collapse
Affiliation(s)
- Lisa Strasser
- Characterization and Comparability Laboratory, National Institute for Bioprocessing Research and Training, Dublin, Ireland
| | - Amy Farrell
- Characterization and Comparability Laboratory, National Institute for Bioprocessing Research and Training, Dublin, Ireland
| | - Jenny T C Ho
- Thermo Fisher Scientific, Hemel Hempstead, United Kingdom
| | | | - Ken Cook
- Thermo Fisher Scientific, Hemel Hempstead, United Kingdom
| | | | - Peter Mowlds
- Thermo Fisher Scientific, Hemel Hempstead, United Kingdom
| | - Rosa Viner
- Thermo Fisher Scientific, San Jose, CA, United States
| | - Barry L Karger
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, United States
| | - Jonathan Bones
- School of Chemical and Bioprocess Engineering, University College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
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
|
35
|
Telu KH, Marupaka R, Andriamaharavo NR, Simón-Manso Y, Liang Y, Mirokhin YA, Bukhari TH, Preston RJ, Kashi L, Kelman Z, Stein SE. Creation and filtering of a recurrent spectral library of CHO cell metabolites and media components. Biotechnol Bioeng 2021; 118:1491-1510. [PMID: 33404064 PMCID: PMC8048470 DOI: 10.1002/bit.27661] [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: 08/22/2020] [Revised: 12/02/2020] [Accepted: 12/13/2020] [Indexed: 02/02/2023]
Abstract
This paper reports the first implementation of a new type of mass spectral library for the analysis of Chinese hamster ovary (CHO) cell metabolites that allows users to quickly identify most compounds in any complex metabolite sample. We also describe an annotation methodology developed to filter out artifacts and low‐quality spectra from recurrent unidentified spectra of metabolites. CHO cells are commonly used to produce biological therapeutics. Metabolic profiles of CHO cells and media can be used to monitor process variability and look for markers that discriminate between batches of product. We have created a comprehensive library of both identified and unidentified metabolites derived from CHO cells that can be used in conjunction with tandem mass spectrometry to identify metabolites. In addition, we present a workflow that can be used for assigning confidence to a NIST MS/MS Library search match based on prior probability of general utility. The goal of our work is to annotate and identify (when possible), all liquid chromatography‐mass spectrometry generated metabolite ions as well as create automatable library building and identification pipelines for use by others in the field.
Collapse
Affiliation(s)
- Kelly H Telu
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Ramesh Marupaka
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Nirina R Andriamaharavo
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Yamil Simón-Manso
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Yuxue Liang
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Yuri A Mirokhin
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Tallat H Bukhari
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Renae J Preston
- Biomolecular Labeling Laboratory, Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology and the University of Maryland, Rockville, Maryland, USA
| | - Lila Kashi
- Biomolecular Labeling Laboratory, Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology and the University of Maryland, Rockville, Maryland, USA
| | - Zvi Kelman
- Biomolecular Labeling Laboratory, Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology and the University of Maryland, Rockville, Maryland, USA
| | - Stephen E Stein
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| |
Collapse
|
36
|
Lin D, Yalamanchili HB, Zhang X, Lewis NE, Alves CS, Groot J, Arnsdorf J, Bjørn SP, Wulff T, Voldborg BG, Zhou Y, Zhang B. CHOmics: A web-based tool for multi-omics data analysis and interactive visualization in CHO cell lines. PLoS Comput Biol 2020; 16:e1008498. [PMID: 33351794 PMCID: PMC7790544 DOI: 10.1371/journal.pcbi.1008498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 01/07/2021] [Accepted: 11/06/2020] [Indexed: 11/18/2022] Open
Abstract
Chinese hamster ovary (CHO) cell lines are widely used in industry for biological drug production. During cell culture development, considerable effort is invested to understand the factors that greatly impact cell growth, specific productivity and product qualities of the biotherapeutics. While high-throughput omics approaches have been increasingly utilized to reveal cellular mechanisms associated with cell line phenotypes and guide process optimization, comprehensive omics data analysis and management have been a challenge. Here we developed CHOmics, a web-based tool for integrative analysis of CHO cell line omics data that provides an interactive visualization of omics analysis outputs and efficient data management. CHOmics has a built-in comprehensive pipeline for RNA sequencing data processing and multi-layer statistical modules to explore relevant genes or pathways. Moreover, advanced functionalities were provided to enable users to customize their analysis and visualize the output systematically and interactively. The tool was also designed with the flexibility to accommodate other types of omics data and thereby enabling multi-omics comparison and visualization at both gene and pathway levels. Collectively, CHOmics is an integrative platform for data analysis, visualization and management with expectations to promote the broader use of omics in CHO cell research.
Collapse
Affiliation(s)
- Dongdong Lin
- Biogen Inc., Cambridge, Massachusetts, United States of America
| | | | - Xinmin Zhang
- Bioinforx Inc., Madison, Wisconsin, United States of America
| | - Nathan E. Lewis
- Departments of Pediatrics and Bioengineering, University of California, San Diego, United States of America
- Novo Nordisk Foundation Center for Bio sustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Joost Groot
- Biogen Inc., Cambridge, Massachusetts, United States of America
| | - Johnny Arnsdorf
- Novo Nordisk Foundation Center for Bio sustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Sara P. Bjørn
- Novo Nordisk Foundation Center for Bio sustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Tune Wulff
- Novo Nordisk Foundation Center for Bio sustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Bjørn G. Voldborg
- Novo Nordisk Foundation Center for Bio sustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Yizhou Zhou
- Biogen Inc., Cambridge, Massachusetts, United States of America
- * E-mail: (YZ); (BZ)
| | - Baohong Zhang
- Biogen Inc., Cambridge, Massachusetts, United States of America
- * E-mail: (YZ); (BZ)
| |
Collapse
|
37
|
O’Brien SA, Hu WS. Cell culture bioprocessing - the road taken and the path forward. Curr Opin Chem Eng 2020; 30:100663. [PMID: 33391982 PMCID: PMC7773285 DOI: 10.1016/j.coche.2020.100663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cell culture processes are used to produce the vast majority of protein therapeutics, valued at over US$180 billion per annum worldwide. For more than a decade now, these processes have become highly productive. To further enhance capital efficiency, there has been an increase in the adoption of disposable apparatus and continuous processing, as well as a greater exploration of in-line sensing, various -omic tools, and cell engineering to enhance process controllability and product quality consistency. These feats in cell culture processing for protein biologics will help accelerate the bioprocess advancements for virus and cell therapy applications.
Collapse
Affiliation(s)
- Sofie A. O’Brien
- Department of Biomedical Engineering and Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455-0132 USA
| | - Wei-Shou Hu
- Department of Biomedical Engineering and Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455-0132 USA
| |
Collapse
|
38
|
O’Flaherty R, Bergin A, Flampouri E, Mota LM, Obaidi I, Quigley A, Xie Y, Butler M. Mammalian cell culture for production of recombinant proteins: A review of the critical steps in their biomanufacturing. Biotechnol Adv 2020; 43:107552. [DOI: 10.1016/j.biotechadv.2020.107552] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 12/28/2022]
|
39
|
Hoang Anh N, Min JE, Kim SJ, Phuoc Long N. Biotherapeutic Products, Cellular Factories, and Multiomics Integration in Metabolic Engineering. ACTA ACUST UNITED AC 2020; 24:621-633. [DOI: 10.1089/omi.2020.0112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jung Eun Min
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Sun Jo Kim
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Nguyen Phuoc Long
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
40
|
Orellana CA, Martínez VS, MacDonald MA, Henry MN, Gillard M, Gray PP, Nielsen LK, Mahler S, Marcellin E. 'Omics driven discoveries of gene targets for apoptosis attenuation in CHO cells. Biotechnol Bioeng 2020; 118:481-490. [PMID: 32865815 DOI: 10.1002/bit.27548] [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: 10/28/2019] [Revised: 07/22/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022]
Abstract
Chinese hamster ovary (CHO) cells are widely used in biopharmaceutical production. Improvements to cell lines and bioprocesses are constantly being explored. One of the major limitations of CHO cell culture is that the cells undergo apoptosis, leading to rapid cell death, which impedes reaching high recombinant protein titres. While several genetic engineering strategies have been successfully employed to reduce apoptosis, there is still room to further enhance CHO cell lines performance. 'Omics analysis is a powerful tool to better understand different phenotypes and for the identification of gene targets for engineering. Here, we present a comprehensive review of previous CHO 'omics studies that revealed changes in the expression of apoptosis-related genes. We highlight targets for genetic engineering that have reduced, or have the potential to reduce, apoptosis or to increase cell proliferation in CHO cells, with the final aim of increasing productivity.
Collapse
Affiliation(s)
- Camila A Orellana
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia.,Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Verónica S Martínez
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| | - Michael A MacDonald
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| | - Matthew N Henry
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| | - Marianne Gillard
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| | - Peter P Gray
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| | - Lars K Nielsen
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia.,Metabolomics Australia, The University of Queensland, Brisbane, Australia.,The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Stephen Mahler
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia
| | - Esteban Marcellin
- ARC Training Centre for Biopharmaceutical Innovation (CBI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Australia.,Metabolomics Australia, The University of Queensland, Brisbane, Australia
| |
Collapse
|
41
|
Identifying metabolic features and engineering targets for productivity improvement in CHO cells by integrated transcriptomics and genome-scale metabolic model. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
42
|
Li SW, Wright M, Healey JF, Hutchinson JM, O’Rourke S, Mesa KA, Lollar P, Berman PW. Gene editing in CHO cells to prevent proteolysis and enhance glycosylation: Production of HIV envelope proteins as vaccine immunogens. PLoS One 2020; 15:e0233866. [PMID: 32470085 PMCID: PMC7259603 DOI: 10.1371/journal.pone.0233866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/13/2020] [Indexed: 01/12/2023] Open
Abstract
Several candidate HIV subunit vaccines based on recombinant envelope (Env) glycoproteins have been advanced into human clinical trials. To facilitate biopharmaceutical production, it is necessary to produce these in CHO (Chinese Hamster Ovary) cells, the cellular substrate used for the manufacturing of most recombinant protein therapeutics. However, previous studies have shown that when recombinant Env proteins from clade B viruses, the major subtype represented in North America, Europe, and other parts of the world, are expressed in CHO cells, they are proteolyzed and lack important glycan-dependent epitopes present on virions. Previously, we identified C1s, a serine protease in the complement pathway, as the endogenous CHO protease responsible for the cleavage of clade B laboratory isolates of -recombinant gp120s (rgp120s) expressed in stable CHO-S cell lines. In this paper, we describe the development of two novel CHOK1 cell lines with the C1s gene inactivated by gene editing, that are suitable for the production of any protein susceptible to C1s proteolysis. One cell line, C1s-/- CHOK1 2.E7, contains a deletion in the C1s gene. The other cell line, C1s-/- MGAT1- CHOK1 1.A1, contains a deletion in both the C1s gene and the MGAT1 gene, which limits glycosylation to mannose-5 or earlier intermediates in the N-linked glycosylation pathway. In addition, we compare the substrate specificity of C1s with thrombin on the cleavage of both rgp120 and human Factor VIII, two recombinant proteins known to undergo unintended proteolysis (clipping) when expressed in CHO cells. Finally, we demonstrate the utility and practicality of the C1s-/- MGAT1- CHOK1 1.A1 cell line for the expression of clinical isolates of clade B Envs from rare individuals that possess broadly neutralizing antibodies and are able to control virus replication without anti-retroviral drugs (elite neutralizer/controller phenotypes). The Envs represent unique HIV vaccine immunogens suitable for further immunogenicity and efficacy studies.
Collapse
Affiliation(s)
- Sophia W. Li
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California, United States of America
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Meredith Wright
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - John F. Healey
- Department of Pediatrics, Emory University, Atlanta, Georgia, United States of America
| | - Jennie M. Hutchinson
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Sara O’Rourke
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Kathryn A. Mesa
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Pete Lollar
- Department of Pediatrics, Emory University, Atlanta, Georgia, United States of America
| | - Phillip W. Berman
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, United States of America
| |
Collapse
|
43
|
Srirangan K, Loignon M, Durocher Y. The use of site-specific recombination and cassette exchange technologies for monoclonal antibody production in Chinese Hamster ovary cells: retrospective analysis and future directions. Crit Rev Biotechnol 2020; 40:833-851. [DOI: 10.1080/07388551.2020.1768043] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Kajan Srirangan
- 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
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
44
|
Alden N, Raju R, McElearney K, Lambropoulos J, Kshirsagar R, Gilbert A, Lee K. Using Metabolomics to Identify Cell Line-Independent Indicators of Growth Inhibition for Chinese Hamster Ovary Cell-based Bioprocesses. Metabolites 2020; 10:metabo10050199. [PMID: 32429145 PMCID: PMC7281457 DOI: 10.3390/metabo10050199] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 12/22/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are widely used for the production of biopharmaceuticals. Efforts to improve productivity through medium design and feeding strategy optimization have focused on preventing the depletion of essential nutrients and managing the accumulation of lactate and ammonia. In addition to ammonia and lactate, many other metabolites accumulate in CHO cell cultures, although their effects remain largely unknown. Elucidating these effects has the potential to further improve the productivity of CHO cell-based bioprocesses. This study used untargeted metabolomics to identify metabolites that accumulate in fed-batch cultures of monoclonal antibody (mAb) producing CHO cells. The metabolomics experiments profiled six cell lines that are derived from two different hosts, produce different mAbs, and exhibit different growth profiles. Comparing the cell lines’ metabolite profiles at different growth stages, we found a strong negative correlation between peak viable cell density (VCD) and a tryptophan metabolite, putatively identified as 5-hydroxyindoleacetaldehyde (5-HIAAld). Amino acid supplementation experiments showed strong growth inhibition of all cell lines by excess tryptophan, which correlated with the accumulation of 5-HIAAld in the culture medium. Prospectively, the approach presented in this study could be used to identify cell line- and host-independent metabolite markers for clone selection and bioprocess development.
Collapse
Affiliation(s)
- Nicholas Alden
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA;
| | - Ravali Raju
- Biogen, 225 Binney St, Cambridge, MA 02142, USA; (R.R.); (K.M.); (J.L.); (R.K.); (A.G.)
| | - Kyle McElearney
- Biogen, 225 Binney St, Cambridge, MA 02142, USA; (R.R.); (K.M.); (J.L.); (R.K.); (A.G.)
| | - James Lambropoulos
- Biogen, 225 Binney St, Cambridge, MA 02142, USA; (R.R.); (K.M.); (J.L.); (R.K.); (A.G.)
| | - Rashmi Kshirsagar
- Biogen, 225 Binney St, Cambridge, MA 02142, USA; (R.R.); (K.M.); (J.L.); (R.K.); (A.G.)
| | - Alan Gilbert
- Biogen, 225 Binney St, Cambridge, MA 02142, USA; (R.R.); (K.M.); (J.L.); (R.K.); (A.G.)
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA;
- Correspondence:
| |
Collapse
|
45
|
Combining lipoic acid to methylene blue reduces the Warburg effect in CHO cells: From TCA cycle activation to enhancing monoclonal antibody production. PLoS One 2020; 15:e0231770. [PMID: 32298377 PMCID: PMC7162497 DOI: 10.1371/journal.pone.0231770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/31/2020] [Indexed: 01/01/2023] Open
Abstract
The Warburg effect, a hallmark of cancer, has recently been identified as a metabolic limitation of Chinese Hamster Ovary (CHO) cells, the primary platform for the production of monoclonal antibodies (mAb). Metabolic engineering approaches, including genetic modifications and feeding strategies, have been attempted to impose the metabolic prevalence of respiration over aerobic glycolysis. Their main objective lies in decreasing lactate production while improving energy efficiency. Although yielding promising increases in productivity, such strategies require long development phases and alter entangled metabolic pathways which singular roles remain unclear. We propose to apply drugs used for the metabolic therapy of cancer to target the Warburg effect at different levels, on CHO cells. The use of α-lipoic acid, a pyruvate dehydrogenase activator, replenished the Krebs cycle through increased anaplerosis but resulted in mitochondrial saturation. The electron shuttle function of a second drug, methylene blue, enhanced the mitochondrial capacity. It pulled on anaplerotic pathways while reducing stress signals and resulted in a 24% increase of the maximum mAb production. Finally, the combination of both drugs proved to be promising for stimulating Krebs cycle activity and mitochondrial respiration. Therefore, drugs used in metabolic therapy are valuable candidates to understand and improve the metabolic limitations of CHO-based bioproduction.
Collapse
|
46
|
Ma X, Zhang L, Zhang L, Wang C, Guo X, Yang Y, Wang L, Li X, Ma N. Validation and identification of reference genes in Chinese hamster ovary cells for Fc-fusion protein production. Exp Biol Med (Maywood) 2020; 245:690-702. [PMID: 32216463 DOI: 10.1177/1535370220914058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
IMPACT STATEMENT In order to reveal potential genotype-phenotype relationship, RT-qPCR reactions are frequently applied which require validated and reliable reference genes. With the investigation on long-term passage and fed-batch cultivation of CHO cells producing an Fc-fusion protein, four new reference genes-Akr1a1, Gpx1, Aprt, and Rps16, were identified from 20 candidates with the aid of geNorm, NormFinder, BestKeeper, and ΔCt programs and methods. This article provided more verified options in reference gene selection in related research on CHO cells.
Collapse
Affiliation(s)
- Xiaonan Ma
- Wuya college of Innovation; College of life science and biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ling Zhang
- Wuya college of Innovation; College of life science and biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Luming Zhang
- Wuya college of Innovation; College of life science and biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chenglong Wang
- Wuya college of Innovation; College of life science and biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaorui Guo
- Wuya college of Innovation; College of life science and biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Yang
- Wuya college of Innovation; College of life science and biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lin Wang
- Wuya college of Innovation; College of life science and biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiangru Li
- Wuya college of Innovation; College of life science and biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ningning Ma
- Wuya college of Innovation; College of life science and biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
47
|
Yagi H, Yagi-Utsumi M, Honda R, Ohta Y, Saito T, Nishio M, Ninagawa S, Suzuki K, Anzai T, Kamiya Y, Aoki K, Nakanishi M, Satoh T, Kato K. Improved secretion of glycoproteins using an N-glycan-restricted passport sequence tag recognized by cargo receptor. Nat Commun 2020; 11:1368. [PMID: 32170195 PMCID: PMC7069976 DOI: 10.1038/s41467-020-15192-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 02/25/2020] [Indexed: 11/09/2022] Open
Abstract
MCFD2 and ERGIC-53, which are the products of causative genes of combined factor V and factor VIII deficiency, form a cargo receptor complex responsible for intracellular transport of these coagulation factors in the early secretory pathway. In this study, using an NMR technique, we successfully identified an MCFD2-binding segment from factor VIII composed of a 10 amino acid sequence that enhances its secretion. This prompted us to examine possible effects of attaching this sequence to recombinant glycoproteins on their secretion. We found that the secretion level of recombinant erythropoietin was significantly increased simply by tagging it with the passport sequence. Our findings not only provide molecular basis for the intracellular trafficking of coagulation factors and their genetic deficiency but also offer a potentially useful tool for increasing the production yields of recombinant glycoproteins of biopharmaceutical interest.
Collapse
Affiliation(s)
- Hirokazu Yagi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Maho Yagi-Utsumi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
- Institute for Molecular Science, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
- School of Physical Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
| | - Rena Honda
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
- Institute for Molecular Science, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
- School of Physical Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
| | - Yusaku Ohta
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
- National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
| | - Taiki Saito
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Miho Nishio
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Satoshi Ninagawa
- Institute for Molecular Science, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
| | - Kousuke Suzuki
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Takahiro Anzai
- Institute for Molecular Science, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
| | - Yukiko Kamiya
- Institute for Molecular Science, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
| | - Kazuhiro Aoki
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
- National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan
| | - Mahito Nakanishi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Central 5, Tsukuba, Ibaraki, 305-8565, Japan
| | - Tadashi Satoh
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Koichi Kato
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan.
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan.
- Institute for Molecular Science, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan.
- School of Physical Science, SOKENDAI (The Graduate University for Advanced Studies), 5-1 Higashiyama, Myodaiji, Okazaki, 444-8787, Japan.
| |
Collapse
|
48
|
Joubert S, Dodelet V, Béliard R, Durocher Y. [Biomanufacturing of monoclonal antibodies]. Med Sci (Paris) 2020; 35:1153-1159. [PMID: 31903930 DOI: 10.1051/medsci/2019219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Antibody-based drugs are an increasingly important part of the therapeutic arsenal against a wide variety of medical conditions. As the number of commercial products and pipeline candidates grows, a crucial issue facing the industry is the current and future state of biomanufacturing. The productivity of the protein expression platforms, along with the performance of the technologies impacting upstream and downstream bioprocessing, are critical factors affecting the cost and time of therapeutic antibody development and commercialization. Cell engineering strategies are being used to improve the production of antibodies and to better control their quality in terms of posttranslational modifications, in particular with regards to their glycosylation state, as this can influence their therapeutic activity. Additionally, the advance of "omics" technologies have recently given rise to new possibilities in improving these expression platforms. We review here the various advances in biomanufacturing essential to the continued growth of the therapeutic antibody market.
Collapse
Affiliation(s)
- Simon Joubert
- Centre de recherche sur les thérapeutiques en santé humaine, Conseil national de recherche du Canada, Montréal, Québec H4P 2R2, Canada
| | - Vincent Dodelet
- Centre de recherche sur les thérapeutiques en santé humaine, Conseil national de recherche du Canada, Montréal, Québec H4P 2R2, Canada
| | - Roland Béliard
- Laboratoires français du fractionnement et des biotechnologies, Les Ulis, Courtaboeuf Cedex, France
| | - Yves Durocher
- Centre de recherche sur les thérapeutiques en santé humaine, Conseil national de recherche du Canada, Montréal, Québec H4P 2R2, Canada - Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| |
Collapse
|
49
|
Huhn SC, Ou Y, Kumar A, Liu R, Du Z. High throughput, efficacious gene editing & genome surveillance in Chinese hamster ovary cells. PLoS One 2019; 14:e0218653. [PMID: 31856197 PMCID: PMC6922373 DOI: 10.1371/journal.pone.0218653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 06/06/2019] [Indexed: 12/26/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are a common tool utilized in bioproduction and directed genome engineering of CHO cells is of great interest to enhance recombinant cell lines. Until recently, this focus has been challenged by a lack of efficacious, high throughput, and low-cost gene editing modalities and screening methods. In this work, we demonstrate an improved method for gene editing in CHO cells using CRISPR RNPs and characterize the endpoints of Cas9 and ZFN mediated genetic engineering. Furthermore, we validate sequence decomposition as a cost effective, rapid, and accurate method for assessing mutants and eliminating non-clonal CHO populations using only capillary sequencing.
Collapse
Affiliation(s)
- S. C. Huhn
- Cell Line Development, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Y. Ou
- Cell Line Development, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - A. Kumar
- Cell Line Development, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - R. Liu
- Cell Line Development, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Z. Du
- Cell Line Development, Merck & Co., Inc., Kenilworth, NJ, United States of America
- * E-mail:
| |
Collapse
|
50
|
Amann T, Schmieder V, Faustrup Kildegaard H, Borth N, Andersen MR. Genetic engineering approaches to improve posttranslational modification of biopharmaceuticals in different production platforms. Biotechnol Bioeng 2019; 116:2778-2796. [PMID: 31237682 DOI: 10.1002/bit.27101] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/27/2019] [Accepted: 06/18/2019] [Indexed: 12/18/2022]
Abstract
The number of approved biopharmaceuticals, where product quality attributes remain of major importance, is increasing steadily. Within the available variety of expression hosts, the production of biopharmaceuticals faces diverse limitations with respect to posttranslational modifications (PTM), while different biopharmaceuticals demand different forms and specifications of PTMs for proper functionality. With the growing toolbox of genetic engineering technologies, it is now possible to address general as well as host- or biopharmaceutical-specific product quality obstacles. In this review, we present diverse expression systems derived from mammalians, bacteria, yeast, plants, and insects as well as available genetic engineering tools. We focus on genes for knockout/knockdown and overexpression for meaningful approaches to improve biopharmaceutical PTMs and discuss their applicability as well as future trends in the field.
Collapse
Affiliation(s)
- Thomas Amann
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Valerie Schmieder
- acib GmbH-Austrian Centre of Industrial Biotechnology, Graz, Austria.,Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Helene Faustrup Kildegaard
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Nicole Borth
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Mikael Rørdam Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|