1
|
Zhou Z“Z, Si Y, Zhang J, Chen K, George A, Kim S, Zhou L, Liu X“M. A Dual-Payload Antibody-Drug Conjugate Targeting CD276/B7-H3 Elicits Cytotoxicity and Immune Activation in Triple-Negative Breast Cancer. Cancer Res 2024; 84:3848-3863. [PMID: 39186778 PMCID: PMC11565169 DOI: 10.1158/0008-5472.can-23-4099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/30/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and heterogeneous disease that often relapses following treatment with standard radiotherapies and cytotoxic chemotherapies. Combination therapies have potential for treating refractory metastatic TNBC. In this study, we aimed to develop an antibody-drug conjugate with dual payloads (DualADC) as a chemoimmunotherapy for TNBC. The overexpression of an immune checkpoint transmembrane CD276 (also known as B7-H3) was associated with angiogenesis, metastasis, and immune tolerance in more than 60% of patients with TNBC. Development of a mAb capable of targeting the extracellular domain of surface CD276 enabled delivery of payloads to tumors, and a platform was established for concurrent conjugation of a traditional cytotoxic payload and an immunoregulating Toll-like receptor 7/8 agonist to the CD276 mAb. The DualADC effectively killed multiple TNBC subtypes, significantly enhanced immune functions in the tumor microenvironment, and reduced tumor burden by up to 90% to 100% in animal studies. Single-cell RNA sequencing, multiplex cytokine analysis, and histology elucidated the impact of treatment on tumor cells and the immune landscape. This study suggests that the developed DualADC could represent a promising targeted chemoimmunotherapy for TNBC. Significance: An anti-CD276 monoclonal antibody conjugated with both a cytotoxic drug and an immune boosting reagent effectively targets triple-negative breast cancer by inducing tumor cell death and stimulating immune cell infiltration.
Collapse
Affiliation(s)
- Zhuoxin “Zora” Zhou
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Yingnan Si
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Jiashuai Zhang
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Kai Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Ashley George
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Seulhee Kim
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Lufang Zhou
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Xiaoguang “Margaret” Liu
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
2
|
Desmurget C, Perilleux A, Souquet J, Borth N, Douet J. Molecular biomarkers identification and applications in CHO bioprocessing. J Biotechnol 2024; 392:11-24. [PMID: 38852681 DOI: 10.1016/j.jbiotec.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Biomarkers are valuable tools in clinical research where they allow to predict susceptibility to diseases, or response to specific treatments. Likewise, biomarkers can be extremely useful in the biomanufacturing of therapeutic proteins. Indeed, constraints such as short timelines and the need to find hyper-productive cells could benefit from a data-driven approach during cell line and process development. Many companies still rely on large screening capacities to develop productive cell lines, but as they reach a limit of production, there is a need to go from empirical to rationale procedures. Similarly, during bioprocessing runs, substrate consumption and metabolism wastes are commonly monitored. None of them possess the ability to predict the culture behavior in the bioreactor. Big data driven approaches are being adapted to the study of industrial mammalian cell lines, enabled by the publication of Chinese hamster and CHO genome assemblies which allowed the use of next-generation sequencing with these cells, as well as continuous proteome and metabolome annotation. However, if these different -omics technologies contributed to the characterization of CHO cells, there is a significant effort remaining to apply this knowledge to biomanufacturing methods. The correlation of a complex phenotype such as high productivity or rapid growth to the presence or expression level of a specific biomarker could save time and effort in the screening of manufacturing cell lines or culture conditions. In this review we will first discuss the different biological molecules that can be identified and quantified in cells, their detection techniques, and associated challenges. We will then review how these markers are used during the different steps of cell line and bioprocess development, and the inherent limitations of this strategy.
Collapse
Affiliation(s)
- Caroline Desmurget
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Arnaud Perilleux
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Jonathan Souquet
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland
| | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Julien Douet
- Merck Biotech Development Center, Ares Trading SA (an affiliate of Merck KGaA, Darmstadt, Germany), Fenil-sur-Corsier, Switzerland.
| |
Collapse
|
3
|
Zhang D, Wickramasinghe SR, Zydney AL, Smelko JP, Loman A, Wheeler A, Qian X. Proteomic analysis of host cell protein fouling during bioreactor harvesting. Biotechnol Prog 2024; 40:e3453. [PMID: 38477450 DOI: 10.1002/btpr.3453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Chinese hamster ovary (CHO) cells are among the most common cell lines used for therapeutic protein production. Membrane fouling during bioreactor harvesting is a major limitation for the downstream purification of therapeutic proteins. Host cell proteins (HCP) are the most challenging impurities during downstream purification processes. The present work focuses on identification of HCP foulants during CHO bioreactor harvesting using reverse asymmetrical commercial membrane BioOptimal™ MF-SL. In order to investigate foulants and fouling behavior during cell clarification, for the first time a novel backwash process was developed to effectively elute almost all the HCP and DNA from the fouled membrane filter. The isoelectric points (pIs) and molecular weights (MWs) of major HCP in the bioreactor harvest and fouled on the membrane were successfully characterized using two-dimensional gel electrophoresis (2D SDS-PAGE). In addition, a total of 8 HCP were identified using matrix-assisted laser desorption/ionization-mass spectroscopy (MALDI-MS). The majority of these HCP are enzymes or associated with exosomes, both of which can form submicron-sized particles which could lead to the plugging of the filters.
Collapse
Affiliation(s)
- Da Zhang
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - S Ranil Wickramasinghe
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Andrew L Zydney
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - John P Smelko
- Biogen, Research Triangle Park, Durham, North Carolina, USA
| | - Abdullah Loman
- Biogen, Research Triangle Park, Durham, North Carolina, USA
| | - April Wheeler
- Asahi Kasei Bioprocess American, Glenview, Illinois, USA
| | - Xianghong Qian
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
4
|
Sulaj E, Schwaigerlehner L, Sandell FL, Dohm JC, Marzban G, Kunert R. Quantitative proteomics reveals cellular responses to individual mAb expression and tunicamycin in CHO cells. Appl Microbiol Biotechnol 2024; 108:381. [PMID: 38896138 PMCID: PMC11186912 DOI: 10.1007/s00253-024-13223-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Chinese hamster ovary (CHO) cells are popular in the pharmaceutical industry for their ability to produce high concentrations of antibodies and their resemblance to human cells in terms of protein glycosylation patterns. Current data indicate the relevance of CHO cells in the biopharmaceutical industry, with a high number of product commendations and a significant market share for monoclonal antibodies. To enhance the production capabilities of CHO cells, a deep understanding of their cellular and molecular composition is crucial. Genome sequencing and proteomic analysis have provided valuable insights into the impact of the bioprocessing conditions, productivity, and product quality. In our investigation, we conducted a comparative analysis of proteomic profiles in high and low monoclonal antibody-producing cell lines and studied the impact of tunicamycin (TM)-induced endoplasmic reticulum (ER) stress. We examined the expression levels of different proteins including unfolded protein response (UPR) target genes by using label-free quantification techniques for protein abundance. Our results show the upregulation of proteins associated with protein folding mechanisms in low producer vs. high producer cell line suggesting a form of ER stress related to specific protein production. Further, Hspa9 and Dnaja3 are notable candidates activated by the mitochondria UPR and play important roles in protein folding processes in mitochondria. We identified significant upregulation of Nedd8 and Lgmn proteins in similar levels which may contribute to UPR stress. Interestingly, the downregulation of Hspa5/Bip and Pdia4 in response to tunicamycin treatment suggests a low-level UPR activation. KEY POINTS: • Proteome profiling of recombinant CHO cells under mild TM treatment. • Identified protein clusters are associated with the unfolded protein response (UPR). • The compared cell lines revealed noticeable disparities in protein expression levels.
Collapse
Affiliation(s)
- Eldi Sulaj
- Department of Biotechnology, Institute of Animal Cell Technology and Systems Biology (IACTSB), BOKU University, Muthgasse 18, 1190, Vienna, Austria
| | - Linda Schwaigerlehner
- Department of Biotechnology, Institute of Animal Cell Technology and Systems Biology (IACTSB), BOKU University, Muthgasse 18, 1190, Vienna, Austria
| | - Felix L Sandell
- Department of Biotechnology, Institute of Computational Biology (ICB), BOKU University, Muthgasse 18, 1190, Vienna, Austria
| | - Juliane C Dohm
- Department of Biotechnology, Institute of Computational Biology (ICB), BOKU University, Muthgasse 18, 1190, Vienna, Austria
| | - Gorji Marzban
- Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), BOKU University, Muthgasse 18, 1190, Vienna, Austria.
| | - Renate Kunert
- Department of Biotechnology, Institute of Animal Cell Technology and Systems Biology (IACTSB), BOKU University, Muthgasse 18, 1190, Vienna, Austria
| |
Collapse
|
5
|
Joseph J, Sandel G, Kulkarni R, Alatrash R, Herrera BB, Jain P. Antibody and Cell-Based Therapies against Virus-Induced Cancers in the Context of HIV/AIDS. Pathogens 2023; 13:14. [PMID: 38251321 PMCID: PMC10821063 DOI: 10.3390/pathogens13010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
Infectious agents, notably viruses, can cause or increase the risk of cancer occurrences. These agents often disrupt normal cellular functions, promote uncontrolled proliferation and growth, and trigger chronic inflammation, leading to cancer. Approximately 20% of all cancer cases in humans are associated with an infectious pathogen. The International Agency for Research on Cancer (IARC) recognizes seven viruses as direct oncogenic agents, including Epstein-Barr Virus (EBV), Kaposi's Sarcoma-associated herpesvirus (KSHV), human T-cell leukemia virus type-1 (HTLV-1), human papilloma virus (HPV), hepatitis C virus (HCV), hepatitis B virus (HBV), and human immunodeficiency virus type 1 (HIV-1). Most viruses linked to increased cancer risk are typically transmitted through contact with contaminated body fluids and high-risk behaviors. The risk of infection can be reduced through vaccinations and routine testing, as well as recognizing and addressing risky behaviors and staying informed about public health concerns. Numerous strategies are currently in pre-clinical phases or undergoing clinical trials for targeting cancers driven by viral infections. Herein, we provide an overview of risk factors associated with increased cancer incidence in people living with HIV (PLWH) as well as other chronic viral infections, and contributing factors such as aging, toxicity from ART, coinfections, and comorbidities. Furthermore, we highlight both antibody- and cell-based strategies directed against virus-induced cancers while also emphasizing approaches aimed at discovering cures or achieving complete remission for affected individuals.
Collapse
Affiliation(s)
- Julie Joseph
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Grace Sandel
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Ratuja Kulkarni
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Reem Alatrash
- Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; (R.A.); (B.B.H.)
- Department of Medicine, Division of Allergy, Immunology and Infectious Diseases, Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Bobby Brooke Herrera
- Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; (R.A.); (B.B.H.)
- Department of Medicine, Division of Allergy, Immunology and Infectious Diseases, Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Pooja Jain
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| |
Collapse
|
6
|
Ihling N, Berg C, Paul R, Munkler LP, Mäkinen MEL, Chotteau V, Büchs J. Scale-down of CHO cell cultivation from shake flasks based on oxygen mass transfer allows application of parallelized, non-invasive, and time-resolved monitoring of the oxygen transfer rate in 48-well microtiter plates. Biotechnol J 2023; 18:e2300053. [PMID: 37424196 DOI: 10.1002/biot.202300053] [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: 02/02/2023] [Revised: 06/23/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Cultivating Chinese hamster ovary (CHO) cells in microtiter plates (MTPs) with time-resolved monitoring of the oxygen transfer rate (OTR) is highly desirable to provide process insights at increased throughput. However, monitoring of the OTR in MTPs has not been demonstrated for CHO cells, yet. Hence, a CHO cultivation process was transferred from shake flasks to MTPs to enable monitoring of the OTR in each individual well of a 48-well MTP. For this, the cultivation of an industrially relevant, antibody-producing cell line was transferred from shake flask to MTP based on the volumetric oxygen mass transfer coefficient (kL a). Culture behavior was well comparable (deviation of the final IgG titer less than 10%). Monitoring of the OTR in 48-well MTPs was then used to derive the cytotoxicity of dimethyl sulfoxide (DMSO) based on a dose-response curve in a single experiment using a second CHO cell line. Logistic fitting of the dose-response curve determined after 100 h was used to determine the DMSO concentration that resulted in a cytotoxicity of 50% (IC50). A DMSO concentration of 2.70% ± 0.25% was determined, which agrees with the IC50 previously determined in shake flasks (2.39% ± 0.1%). Non-invasive, parallelized, and time-resolved monitoring of the OTR of CHO cells in MTPs was demonstrated and offers excellent potential to speed up process development and assess cytotoxicity.
Collapse
Affiliation(s)
- Nina Ihling
- AVT - Biochemical Engineering, RWTH Aachen University, Aachen, Germany
| | - Christoph Berg
- AVT - Biochemical Engineering, RWTH Aachen University, Aachen, Germany
| | - Richard Paul
- AVT - Biochemical Engineering, RWTH Aachen University, Aachen, Germany
| | | | - Meeri E-L Mäkinen
- KTH Royal Institute of Technology, Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
| | - Veronique Chotteau
- KTH Royal Institute of Technology, Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
- AdBIOPRO, Competence Centre for Advanced BioProduction by Continuous Processing, KTH, Stockholm, Sweden
| | - Jochen Büchs
- AVT - Biochemical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
7
|
Nguyen M, Zimmer A. A reflection on the improvement of Chinese Hamster ovary cell-based bioprocesses through advances in proteomic techniques. Biotechnol Adv 2023; 65:108141. [PMID: 37001570 DOI: 10.1016/j.biotechadv.2023.108141] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 03/05/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
Chinese hamster ovary (CHO) cells are the preferred mammalian host for the large-scale production of recombinant proteins in the biopharmaceutical industry. Research endeavors have been directed to the optimization of CHO-based bioprocesses to increase protein quantity and quality, often in an empirical manner. To provide a rationale for those achievements, a myriad of CHO proteomic studies has arisen in recent decades. This review gives an overview of significant advances in LC-MS-based proteomics and sheds light on CHO proteomic studies, with a particular focus on CHO cells with superior bioprocessing phenotypes (growth, viability, titer, productivity and cQA), that have exploited novel proteomic or sub-omic techniques. These proteomic findings expand the current knowledge and understanding about the underlying protein clusters, protein regulatory networks and biological pathways governing such phenotypic changes. The proteomic studies, highlighted herein, will help in the targeted modulation of these cell factories to the desired needs.
Collapse
|
8
|
Spatial Proteomics Reveals Differences in the Cellular Architecture of Antibody-Producing CHO and Plasma Cell-Derived Cells. Mol Cell Proteomics 2022; 21:100278. [PMID: 35934186 PMCID: PMC9562429 DOI: 10.1016/j.mcpro.2022.100278] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 01/18/2023] Open
Abstract
Most of the recombinant biotherapeutics employed today to combat severe illnesses, for example, various types of cancer or autoimmune diseases, are produced by Chinese hamster ovary (CHO) cells. To meet the growing demand of these pharmaceuticals, CHO cells are under constant development in order to enhance their stability and productivity. The last decades saw a shift from empirical cell line optimization toward rational cell engineering using a growing number of large omics datasets to alter cell physiology on various levels. Especially proteomics workflows reached new levels in proteome coverage and data quality because of advances in high-resolution mass spectrometry instrumentation. One type of workflow concentrates on spatial proteomics by usage of subcellular fractionation of organelles with subsequent shotgun mass spectrometry proteomics and machine learning algorithms to determine the subcellular localization of large portions of the cellular proteome at a certain time point. Here, we present the first subcellular spatial proteome of a CHO-K1 cell line producing high titers of recombinant antibody in comparison to the spatial proteome of an antibody-producing plasma cell-derived myeloma cell line. Both cell lines show colocalization of immunoglobulin G chains with chaperones and proteins associated in protein glycosylation within the endoplasmic reticulum compartment. However, we report differences in the localization of proteins associated to vesicle-mediated transport, transcription, and translation, which may affect antibody production in both cell lines. Furthermore, pairing subcellular localization data with protein expression data revealed elevated protein masses for organelles in the secretory pathway in plasma cell-derived MPC-11 (Merwin plasma cell tumor-11) cells. Our study highlights the potential of subcellular spatial proteomics combined with protein expression as potent workflow to identify characteristics of highly efficient recombinant protein-expressing cell lines. Data are available via ProteomeXchange with identifier PXD029115.
Collapse
|
9
|
Tanemura H, Masuda K, Okumura T, Takagi E, Kajihara D, Kakihara H, Nonaka K, Ushioda R. Development of a stable antibody production system utilizing an Hspa5 promoter in CHO cells. Sci Rep 2022; 12:7239. [PMID: 35610229 PMCID: PMC9130236 DOI: 10.1038/s41598-022-11342-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/19/2022] [Indexed: 11/28/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are widely used for manufacturing antibody drugs. We attempted to clone a novel high-expression promoter for producing monoclonal antibodies (mAbs) based on transcriptome analysis to enhance the transcriptional abundance of mAb genes. The efficacy of conventional promoters such as CMV and hEF1α decrease in the latter phase of fed-batch cell culture. To overcome this, we screened genes whose expression was maintained or increased throughout the culture period. Since CHO cells have diverse genetic expression depending on the selected clone and culture medium, transcriptome analysis was performed on multiple clones and culture media anticipated to be used in mAb manufacturing. We thus acquired the Hspa5 promoter as a novel high-expression promoter, which uniquely enables mAb productivity per cell to improve late in the culture period. Productivity also improved for various IgG subclasses under Hspa5 promoter control, indicating this promoter’s potential universal value for mAb production. Finally, it was suggested that mAb production with this promoter is correlated with the transcription levels of endoplasmic reticulum stress-related genes. Therefore, mAb production utilizing the Hspa5 promoter might be a new method for maintaining protein homeostasis and achieving stable expression of introduced mAb genes during fed-batch culture.
Collapse
Affiliation(s)
- Hiroki Tanemura
- Biologics Technology Research Laboratories Biologics Division, Daiichi Sankyo Co., Ltd., 2716-1, Aza Kurakake, Oaza Akaiwa, Chiyoda-machi, Oura-gun, Gunma, 370-0503, Japan
| | - Kenji Masuda
- Biologics Technology Research Laboratories Biologics Division, Daiichi Sankyo Co., Ltd., 2716-1, Aza Kurakake, Oaza Akaiwa, Chiyoda-machi, Oura-gun, Gunma, 370-0503, Japan
| | - Takeshi Okumura
- Biologics Technology Research Laboratories Biologics Division, Daiichi Sankyo Co., Ltd., 2716-1, Aza Kurakake, Oaza Akaiwa, Chiyoda-machi, Oura-gun, Gunma, 370-0503, Japan
| | - Eri Takagi
- Biologics Technology Research Laboratories Biologics Division, Daiichi Sankyo Co., Ltd., 2716-1, Aza Kurakake, Oaza Akaiwa, Chiyoda-machi, Oura-gun, Gunma, 370-0503, Japan
| | - Daisuke Kajihara
- Biologics Technology Research Laboratories Biologics Division, Daiichi Sankyo Co., Ltd., 2716-1, Aza Kurakake, Oaza Akaiwa, Chiyoda-machi, Oura-gun, Gunma, 370-0503, Japan
| | - Hirofumi Kakihara
- Biologics Technology Research Laboratories Biologics Division, Daiichi Sankyo Co., Ltd., 2716-1, Aza Kurakake, Oaza Akaiwa, Chiyoda-machi, Oura-gun, Gunma, 370-0503, Japan
| | - Koichi Nonaka
- Biologics Technology Research Laboratories Biologics Division, Daiichi Sankyo Co., Ltd., 2716-1, Aza Kurakake, Oaza Akaiwa, Chiyoda-machi, Oura-gun, Gunma, 370-0503, Japan
| | - Ryo Ushioda
- Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-ku, Kyoto City, 603-8555, Japan. .,Institute for Protein Dynamics, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-ku, Kyoto City, 603-8555, Japan.
| |
Collapse
|
10
|
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]
|
11
|
Insights into the Impact of Rosmarinic Acid on CHO Cell Culture Improvement through Transcriptomics Analysis. Processes (Basel) 2022. [DOI: 10.3390/pr10030533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The use of antioxidants in Chinese hamster ovary (CHO) cell cultures to improve monoclonal antibody production has been a topic of great interest. Nevertheless, the antioxidants do not have consistent benefits of production improvement, which might be cell line specific and/or process specific. In this work, we investigated how treatment with the antioxidant rosmarinic acid (RA) improved cell growth and titer in CHO cell cultures using transcriptomics. In particular, transcriptomics analysis indicated that RA treatment modified gene expression and strongly affected the MAPK and PI3K/Akt signaling pathways, which regulate cell survival and cell death. Moreover, it was observed that these signaling pathways, which had been identified to be up-regulated on day 2 and day 6 by RA, were also up-regulated over time (from initial growth phase day 2 to slow growth or protein production phase day 6) in both conditions. In summary, this transcriptomics analysis provides insights into the role of the antioxidant RA in industrial cell culture processes. The current study also represents an example in the industry of how omics can be applied to gain an in-depth understanding of CHO cell biology and to identify critical pathways that can contribute to cell culture process improvement and cell line engineering.
Collapse
|
12
|
Park SY, Egan S, Cura AJ, Aron KL, Xu X, Zheng M, Borys M, Ghose S, Li Z, Lee K. Untargeted proteomics reveals upregulation of stress response pathways during CHO-based monoclonal antibody manufacturing process leading to disulfide bond reduction. MAbs 2021; 13:1963094. [PMID: 34424810 PMCID: PMC8386704 DOI: 10.1080/19420862.2021.1963094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Monoclonal antibody (mAb) interchain disulfide bond reduction can cause a loss of function and negatively impact the therapeutic’s efficacy and safety. Disulfide bond reduction has been observed at various stages during the manufacturing process, including processing of the harvested material. The factors and mechanisms driving this phenomenon are not fully understood. In this study, we examined the host cell proteome as a potential factor affecting the susceptibility of a mAb to disulfide bond reduction in the harvested cell culture fluid (HCCF). We used untargeted liquid-chromatography-mass spectrometry-based proteomics experiments in conjunction with a semi-automated protein identification workflow to systematically compare Chinese hamster ovary (CHO) cell protein abundances between bioreactor conditions that result in reduction-susceptible and reduction-free HCCF. Although the growth profiles and antibody titers of these two bioreactor conditions were indistinguishable, we observed broad differences in host cell protein (HCP) expression. We found significant differences in the abundance of glycolytic enzymes, key protein reductases, and antioxidant defense enzymes. Multivariate analysis of the proteomics data determined that upregulation of stress-inducible endoplasmic reticulum (ER) and other chaperone proteins is a discriminatory characteristic of reduction-susceptible HCP profiles. Overall, these results suggest that stress response pathways activated during bioreactor culture increase the reduction-susceptibility of HCCF. Consequently, these pathways could be valuable targets for optimizing culture conditions to improve protein quality.
Collapse
Affiliation(s)
- Seo-Young Park
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, USA.,School of Chemical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Susan Egan
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Anthony J Cura
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Kathryn L Aron
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Xuankuo Xu
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Mengyuan Zheng
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Michael Borys
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Sanchayita Ghose
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Zhengjian Li
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, USA
| |
Collapse
|
13
|
Samoudi M, Masson HO, Kuo CC, Robinson CM, Lewis NE. From omics to Cellular mechanisms in mammalian cell factory development. Curr Opin Chem Eng 2021; 32:100688. [PMID: 37475722 PMCID: PMC10357924 DOI: 10.1016/j.coche.2021.100688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Mammalian cells have been used widely as biopharmaceutical cell factories due to their ability to make complex biotherapeutic proteins with human-compatible modifications. However, their application for some products has been hampered by low protein yields. Numerous studies have aimed to characterize cellular bottlenecks in the hope of boosting protein productivity, but the complexity of the underlying pathways and the diversity of the modifications have complicated cell engineering when relying solely on traditional methodologies. Incorporating omics-based and systems approaches into cell engineering can provide valuable insights into desirable phenotypes of cell factories. Here, we discuss cell engineering strategies for enhancing protein productivity in mammalian cell factories, particularly CHO and HEK293, and the opportunities and limitations of the genome-wide screening and multi-omics approaches for guiding cell engineering. Systems biology strategies will also be discussed to show how they refine our understanding of the cellular mechanisms which will aid in effective engineering strategies.
Collapse
Affiliation(s)
- Mojtaba Samoudi
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Helen O. Masson
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Chih-Chung Kuo
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Caressa M Robinson
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
- National Biologics Facility, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
14
|
Whitt J, Hong WS, Telange RR, Lin CP, Bibb J, Beebe DJ, Chen H, Jaskula-Sztul R. Non-toxic fragment of botulinum neurotoxin type A and monomethyl auristatin E conjugate for targeted therapy for neuroendocrine tumors. Cancer Gene Ther 2020; 27:898-909. [PMID: 32029905 DOI: 10.1038/s41417-020-0167-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 11/27/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Surgical resection is the only cure for neuroendocrine tumors (NETs). However, widespread metastases have already occured by the time of initial diagnosis in many cases making complete surgical removal impossible. We developed a recombinant heavy-chain receptor binding domain (rHCR) of botulinum neurotoxin type A that can specifically target synaptic vesicle 2 (SV2), a surface receptor abundantly expressed in multiple neuroendocrine tumors. Expression of neuroendocrine differentiation markers chromogranin A (CgA) and achaete-scute complex 1 (ASCL1) were signficantly reduced when treated with rHCR. rHCR conjugated to the antimitotic agent monomethyl auristatin E (MMAE) significantly suppressed proliferation of pancreatic carcinoid (BON) and medullary thyroid cancer cells (MZ) at concentrations of 500 and 300 nM respectively, while no growth suppression was observed in pulmonary fibroblasts and cortical neuron control cell lines. In vivo, rHCR-MMAE significantly reduced tumor volume in mouse xenografts with no observed adverse effects. These data suggest recombinant HCR (rHCR) of BoNT/A preferentially targets neuroendocrine cancer without the neurotoxicity of the full BoNT/A and that SV2 is a specific and promising target for delivering drugs to neuroendocrine tumors.
Collapse
Affiliation(s)
- Jason Whitt
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Won S Hong
- Pathology and Laboratory Medicine and Biomedical Engineering, UW-Madison, Madison, WI, USA
| | - Rahul R Telange
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chee Paul Lin
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James Bibb
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David J Beebe
- Pathology and Laboratory Medicine and Biomedical Engineering, UW-Madison, Madison, WI, USA
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | | |
Collapse
|
15
|
Sim KH, Liu LCY, Tan HT, Tan K, Ng D, Zhang W, Yang Y, Tate S, Bi X. A comprehensive CHO SWATH-MS spectral library for robust quantitative profiling of 10,000 proteins. Sci Data 2020; 7:263. [PMID: 32782267 PMCID: PMC7419519 DOI: 10.1038/s41597-020-00594-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/06/2020] [Indexed: 01/08/2023] Open
Abstract
Sequential window acquisition of all theoretical fragment-ion spectra (SWATH) is a data-independent acquisition (DIA) strategy that requires a specific spectral library to generate unbiased and consistent quantitative data matrices of all peptides. SWATH-MS is a promising approach for in-depth proteomic profiling of Chinese hamster Ovary (CHO) cell lines, improving mechanistic understanding of process optimization, and real-time monitoring of process parameters in biologics R&D and manufacturing. However, no spectral library for CHO cells is publicly available. Here we present a comprehensive CHO global spectral library to measure the abundance of more than 10,000 proteins consisting of 199,102 identified peptides from a CHO-K1 cell proteome. The robustness, accuracy and consistency of the spectral library were validated for high confidence in protein identification and reproducible quantification in different CHO-derived cell lines, instrumental setups and downstream processing samples. The availability of a comprehensive SWATH CHO global spectral library will facilitate detailed characterization of upstream and downstream processes, as well as quality by design (QbD) in biomanufacturing. The data have been deposited to ProteomeXchange (PXD016047).
Collapse
Affiliation(s)
- Kae Hwan Sim
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Lillian Chia-Yi Liu
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Hwee Tong Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Kelly Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Daniel Ng
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Wei Zhang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | | | - Xuezhi Bi
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore.
- Duke-NUS Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
16
|
Ou J, Bao T, Ernst P, Si Y, Prabhu SD, Wu H, Zhang J(J, Zhou L, Yang ST, Liu X(M. Intracellular metabolism analysis of Clostridium cellulovorans via modeling integrating proteomics, metabolomics and fermentation. Process Biochem 2020. [DOI: 10.1016/j.procbio.2019.10.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
17
|
Lakshmanan M, Kok YJ, Lee AP, Kyriakopoulos S, Lim HL, Teo G, Poh SL, Tang WQ, Hong J, Tan AH, Bi X, Ho YS, Zhang P, Ng SK, Lee D. Multi‐omics profiling of CHO parental hosts reveals cell line‐specific variations in bioprocessing traits. Biotechnol Bioeng 2019; 116:2117-2129. [DOI: 10.1002/bit.27014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/03/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Meiyappan Lakshmanan
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Yee Jiun Kok
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Alison P. Lee
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Sarantos Kyriakopoulos
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Hsueh Lee Lim
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Gavin Teo
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Swan Li Poh
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Wen Qin Tang
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Jongkwang Hong
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Andy Hee‐Meng Tan
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Xuezhi Bi
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Ying Swan Ho
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Peiqing Zhang
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Say Kong Ng
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
| | - Dong‐Yup Lee
- Bioprocessing Technology InstituteAgency for Science, Technology and Research (A*STAR) Singapore
- School of Chemical EngineeringSungkyunkwan UniversitySuwon Republic of Korea
| |
Collapse
|
18
|
|
19
|
Bioprocess development of antibody-drug conjugate production for cancer treatment. PLoS One 2018; 13:e0206246. [PMID: 30352095 PMCID: PMC6198984 DOI: 10.1371/journal.pone.0206246] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/09/2018] [Indexed: 11/19/2022] Open
Abstract
Antibody-drug conjugate (ADC) is a class of targeted cancer therapies that combine the advantages of monoclonal antibody (mAb)'s specific targeting and chemotherapy's potent cytotoxicity. The therapeutic effect of ADC is significantly affected by its bioproduction process. This study aims to develop an effective ADC production process using anti-HER2 mAb-drug as a model therapeutic. First, a high titer (>2 g/L) of mAb was produced by Chinese hamster ovary cells from fed-batch cell culture. Both live-cell confocal microscopy imaging and flow cytometry analysis demonstrated that the produced mAb and ADC had strong and specific binding to HER2+ cell line BT474. Second, various conjugation conditions of mAb and drug, including linker selection, ratio of drug and mAb, and conjugation approaches, were investigated to improve the production yield and product quality. Finally, the ADC structure and biological quality were evaluated by SDS-PAGE and anti-breast cancer toxicity study, respectively. The ADC with integral molecular structure and high cytotoxicity (IC50 of 1.95 nM) was produced using the optimized production process. The robust bioproduction process could guide the development of ADC-based biopharmaceuticals.
Collapse
|
20
|
Háda V, Bagdi A, Bihari Z, Timári SB, Fizil Á, Szántay C. Recent advancements, challenges, and practical considerations in the mass spectrometry-based analytics of protein biotherapeutics: A viewpoint from the biosimilar industry. J Pharm Biomed Anal 2018; 161:214-238. [PMID: 30205300 DOI: 10.1016/j.jpba.2018.08.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 01/22/2023]
Abstract
The extensive analytical characterization of protein biotherapeutics, especially of biosimilars, is a critical part of the product development and registration. High-resolution mass spectrometry became the primary analytical tool used for the structural characterization of biotherapeutics. Its high instrumental sensitivity and methodological versatility made it possible to use this technique to characterize both the primary and higher-order structure of these proteins. However, even by using high-end instrumentation, analysts face several challenges with regard to how to cope with industrial and regulatory requirements, that is, how to obtain accurate and reliable analytical data in a time- and cost-efficient way. New sample preparation approaches, measurement techniques and data evaluation strategies are available to meet those requirements. The practical considerations of these methods are discussed in the present review article focusing on hot topics, such as reliable and efficient sequencing strategies, minimization of artefact formation during sample preparation, quantitative peptide mapping, the potential of multi-attribute methodology, the increasing role of mass spectrometry in higher-order structure characterization and the challenges of MS-based identification of host cell proteins. On the basis of the opportunities in new instrumental techniques, methodological advancements and software-driven data evaluation approaches, for the future one can envision an even wider application area for mass spectrometry in the biopharmaceutical industry.
Collapse
Affiliation(s)
- Viktor Háda
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary.
| | - Attila Bagdi
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary
| | - Zsolt Bihari
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary
| | | | - Ádám Fizil
- Analytical Department of Biotechnology, Gedeon Richter Plc, Hungary
| | - Csaba Szántay
- Spectroscopic Research Department, Gedeon Richter Plc, Hungary.
| |
Collapse
|
21
|
Singh A, Kildegaard HF, Andersen MR. An Online Compendium of CHO RNA-Seq Data Allows Identification of CHO Cell Line-Specific Transcriptomic Signatures. Biotechnol J 2018; 13:e1800070. [DOI: 10.1002/biot.201800070] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/16/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Ankita Singh
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark; 2800 Kgs. Lyngby Denmark
- Department of Biotechnology and Biomedicine, Technical University of Denmark; 2800 Kgs. Lyngby Denmark
| | - Helene F. Kildegaard
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark; 2800 Kgs. Lyngby Denmark
| | - Mikael R. Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark; 2800 Kgs. Lyngby Denmark
| |
Collapse
|
22
|
Torkashvand F, Mahboudi F, Vossoughi M, Fatemi E, Moosavi Basri SM, Heydari A, Vaziri B. Quantitative Proteomic Analysis of Cellular Responses to a Designed Amino Acid Feed in a Monoclonal Antibody
Producing Chinese Hamster Ovary Cell Line. IRANIAN BIOMEDICAL JOURNAL 2018. [PMID: 29678103 PMCID: PMC6305810 DOI: 10.29252/.22.6.385] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Chinese hamster ovary (CHO) cell line is considered as the most common cell line in the biopharmaceutical industry because of its capability in performing efficient post-translational modifications and producing the recombinant proteins, which are similar to natural human proteins. The optimization of the upstream process via different feed strategies has a great impact on the target molecule expression and yield. Methods: To determine and understand the molecular events beneath the feed effects on the CHO cell, a label-free quantitative proteomic analysis was applied. The proteome changes followed by the addition of a designed amino acid feed to the monoclonal antibody producing CHO cell line culture medium were investigated. Results: The glutathione synthesis, the negative regulation of the programmed cell death, proteasomal catabolic process, and the endosomal transport pathway were up-regulated in the group fed with a designed amino acid feed compared to the control group. Conclusion: Our findings could be helpful to identify new targets for metabolic engineering.
Collapse
Affiliation(s)
- Fatemeh Torkashvand
- Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fereidoun Mahboudi
- Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Manouchehr Vossoughi
- Department of Chemical and Petroleum Engineering, Biochemical and Bioenvironmental Research Center Sharif University of Technology, Tehran, Iran
| | - Elnaz Fatemi
- Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Masoud Moosavi Basri
- Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Amir Heydari
- Department of Chemical Engineering, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Behrouz Vaziri
- Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|