1
|
Shin SW, Kim SH, Gasselin A, Lee GM, Lee JS. Comprehensive genome-scale CRISPR knockout screening of CHO cells. Sci Data 2025; 12:71. [PMID: 39814846 PMCID: PMC11735622 DOI: 10.1038/s41597-025-04438-6] [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: 05/31/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
Chinese hamster ovary (CHO) cells play a pivotal role in the production of recombinant therapeutics. In the present study, we conducted a genome-scale pooled CRISPR knockout (KO) screening using a virus-free, recombinase-mediated cassette exchange-based platform in CHO-K1 host and CHO-K1 derived recombinant cells. Genome-wide guide RNA (gRNA) amplicon sequencing data were generated from cell libraries, as well as short- and long-term KO libraries, and validated through phenotypic assessment and gRNA read count distribution. Additionally, we obtained gRNA amplicon sequencing data from the highly productive recombinant cell populations. By analyzing these datasets, essential genes involved in cell fitness as well as functional target genes associated with therapeutic protein production can be identified. Collectively, our next-generation sequencing datasets, derived from a robust and reliable CRISPR screening method, provide valuable insights into CHO genomic functions, advancing the development of next-generation CHO factories.
Collapse
Affiliation(s)
- Sung Wook Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Su Hyun Kim
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Aghiles Gasselin
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| | - Jae Seong Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
2
|
Boukari I, Rourou S, Bouzazi D, Essafi-Benkhadir K, Kallel H. Strategies for improving expression of recombinant human chorionic gonadotropin in Chinese Hamster Ovary (CHO) cells. Protein Expr Purif 2025; 225:106596. [PMID: 39218246 DOI: 10.1016/j.pep.2024.106596] [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: 06/17/2024] [Revised: 08/14/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Optimizations of the gene expression cassette combined with the selection of an appropriate signal peptide are important factors that must be considered to enhance heterologous protein expression in Chinese Hamster Ovary (CHO) cells. In this study, we investigated the effectiveness of different signal peptides on the production of recombinant human chorionic gonadotropin (r-hCG) in CHO-K1 cells. Four optimized expression constructs containing four promising signal peptides were stably transfected into CHO-K1 cells. The generated CHO-K1 stable pool was then evaluated for r-hCG protein production. Interestingly, human serum albumin and human interleukin-2 signal peptides exhibited relatively greater extracellular secretion of the r-hCG with an average yield of (16.59 ± 0.02 μg/ml) and (14.80 ± 0.13 μg/ml) respectively compared to the native and murine IgGκ light chain signal peptides. The stably transfected CHO pool was further used as the cell substrate to develop an optimized upstream process followed by a downstream phase of the r-hCG. Finally, the biological activity of the purified r-hCG was assessed using in vitro bioassays. The combined data highlight that the choice of signal peptide can be imperative to ensure an optimal secretion of a recombinant protein in CHO cells. In addition, the stable pool technology was a viable approach for the production of biologically active r-hCG at a research scale with acceptable bioprocess performances and consistent product quality.
Collapse
Affiliation(s)
- Iheb Boukari
- Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, LR16IPT01, Biotechnology Development Group, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia; Faculty of Sciences of Bizerte, Carthage University, 7021, Bizerte, Tunisia.
| | - Samia Rourou
- Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, LR16IPT01, Biotechnology Development Group, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia
| | - Dorsaf Bouzazi
- Plateforme de Physiologie et Physiopathologie Cardiovasculaires, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia
| | - Khadija Essafi-Benkhadir
- Laboratory of Molecular Epidemiology and Experimental Pathology, LR16IPT04, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia
| | - Héla Kallel
- Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, LR16IPT01, Biotechnology Development Group, Institut Pasteur de Tunis, University of Tunis El Manar, 1002, Tunis, Tunisia; Univercells SA, Belgium
| |
Collapse
|
3
|
Pérez-Rubio P, Romero EL, Cervera L, Gòdia F, Nielsen LK, Lavado-García J. Systematic insights into cell density-dependent transcriptional responses upon medium replacements. Biomed Pharmacother 2024; 181:117640. [PMID: 39486366 DOI: 10.1016/j.biopha.2024.117640] [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: 08/18/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Understanding the molecular mechanisms governing transfection efficiency and particle secretion in high cell density cultures is critical to overcome the cell density effect upon transient gene expression. The effect of different conditioned media in HEK293 transcriptome at low and high cell density is explored. A systematic pair-wise comparative study was performed to shed light on the effect on previous phenotypical characteristics of different media conditions: fresh, exhausted and media depleted from extracellular vesicles (EVs) as well as associated proteins and RNAs. The obtained results suggest that restorative effects observed in transfection efficiency when employing EV-depleted media may arise predominantly from physicochemical alterations rather than cellular processes. A significant downregulation of genes associated with nucleocytoplasmic transport for the conditions involving the use of exhausted or EV-depleted media was observed. Moreover, upregulation of histone-related genes in EV-depleted media suggest a role for histone signaling in response to cellular stress or growth limitations, thereby highlighting the potential of manipulating histone levels as a promising strategy to enhance transient transfection. It was also corroborated that the accumulation of extracellular matrix proteins upon cell growth may inhibit transfection by an already-known competitive effect between cell membrane-bound and free proteoglycans. Proteomic characterization of EV-depleted media further unveiled enrichment of pathways associated with infection response and double-strand DNA breaks, suggesting that HEK293 cells undergo an induced infection-like state that disrupts cellular processes. Importantly, the study reveals that EV-depleted media stimulates virion release pathways underscoring the complex interplay between extracellular vesicles and viral budding.
Collapse
Affiliation(s)
- Pol Pérez-Rubio
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain.
| | - Elianet Lorenzo Romero
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - Laura Cervera
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - Francesc Gòdia
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - Lars Keld Nielsen
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby 2800, Denmark; Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Australia
| | - Jesús Lavado-García
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby 2800, Denmark.
| |
Collapse
|
4
|
Sun T, Zhang Y, Liang H, Fang W, Qian Z, Tan KW, Li J, Zheng X, Fang M, Zhou H, Zhou W, Zhang S. Establishment of a semi-continuous scale-down clone screening model for intensified perfusion culture. Biotechnol Lett 2024; 46:1085-1093. [PMID: 39066960 DOI: 10.1007/s10529-024-03512-0] [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: 03/26/2024] [Revised: 07/12/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE Perfusion cultures have been extensively used in the biotechnology industry to achieve high yields of recombinant products, especially those with stability issue. The WuXiUP™ platform represents a novel intensified perfusion that can achieve ultra-high productivity. This study describes a representative scale-down 24-deep well plate (24-DWP) cell culture model for intensified perfusion clone screening. METHODS Clonal cell lines were expanded and evaluated in 24-DWP semi-continuous culture. Cell were sampled and counted daily with the aid of an automated liquid handler and high-throughput cell counter. To mimic perfusion culture, 24-DWP plates were spun down and resuspended with fresh medium daily. Top clones were ranked based on growth profiles and productivities. The best performing clones were evaluated on bioreactors. RESULTS The selected clones achieved volumetric productivity (Pv) up to 5 g/L/day when expressing a monoclonal antibody, with the accumulative harvest Pv exceeding 60 g/L in a 21-day cell culture. Product quality attributes of clones cultured in 24-DWP were comparable with those from bioreactors. A high seeding strategy further shortened the clone screening timeline. CONCLUSION In this study, a 24-DWP semi-continuous scale-down model was successfully developed to screen for cell lines suitable for intensified perfusion culture.
Collapse
Affiliation(s)
- Tao Sun
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Yu Zhang
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Hengrui Liang
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Wenjing Fang
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Zichen Qian
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Kee Wee Tan
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Junjie Li
- Bioprocess Research & Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Xiang Zheng
- Bioprocess Research & Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Mingyue Fang
- Bioprocess Research & Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Hang Zhou
- Bioprocess Research & Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Weichang Zhou
- Biologics Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Sam Zhang
- Cell Line Development, WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China.
| |
Collapse
|
5
|
Sookhoo JRV, Schiffman Z, Ambagala A, Kobasa D, Pardee K, Babiuk S. Protein Expression Platforms and the Challenges of Viral Antigen Production. Vaccines (Basel) 2024; 12:1344. [PMID: 39772006 PMCID: PMC11680109 DOI: 10.3390/vaccines12121344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Several protein expression platforms exist for a wide variety of biopharmaceutical needs. A substantial proportion of research and development into protein expression platforms and their optimization since the mid-1900s is a result of the production of viral antigens for use in subunit vaccine research. This review discusses the seven most popular forms of expression systems used in the past decade-bacterial, insect, mammalian, yeast, algal, plant and cell-free systems-in terms of advantages, uses and limitations for viral antigen production in the context of subunit vaccine research. Post-translational modifications, immunogenicity, efficacy, complexity, scalability and the cost of production are major points discussed. Examples of licenced and experimental vaccines are included along with images which summarize the processes involved.
Collapse
Affiliation(s)
- Jamie R. V. Sookhoo
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3R2, Canada; (J.R.V.S.); (A.A.)
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Zachary Schiffman
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (Z.S.); (D.K.)
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Aruna Ambagala
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3R2, Canada; (J.R.V.S.); (A.A.)
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (Z.S.); (D.K.)
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Keith Pardee
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada;
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3R2, Canada; (J.R.V.S.); (A.A.)
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
6
|
Prabhala SV, Marshall B, Galiardi J, Fan Y, Creamer E, Wood DW. Highly selective split intein method for efficient separation and purification of recombinant therapeutic proteins from mammalian cell culture fluid. J Chromatogr A 2024; 1736:465430. [PMID: 39405639 PMCID: PMC11533640 DOI: 10.1016/j.chroma.2024.465430] [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: 06/21/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
Biologics and vaccines have been successfully developed over the last few decades to treat many diseases. Each of these drugs must be highly purified for clinical use. Monoclonal antibodies (mAbs), the dominant therapeutic modality on the market, can be easily purified using the standard Protein A affinity platform. However, no generally applicable affinity platforms are available for the manufacture of other therapeutic proteins for clinical use. Thus, multicolumn chromatography processes for widely being used for product purification. These processes demand significant optimization to meet desired product quality attributes, where each step also decreases final yields. In this work, we demonstrate the novel self-removing iCapTag™ affinity tag, which provides a new platform for capturing, concentrating, and purifying recombinant proteins. Importantly, this system provides a tagless target protein, which is suitable for research and clinical use, where the only requirement for tag removal is a small change in buffer pH. No additional proteins, reagents or cofactors are required. We also present case studies demonstrating the use of iCapTag™ for highly efficient purification of untagged interferon alpha 2b, the ML39 single chain variable fragment (scFv), and the receptor binding domain (RBD) of SARS-CoV-2 spike protein. These proteins were expressed and secreted by Expi293 cells with the self-removing tag fused to their N-terminus. We were able to obtain highly pure (> 99 %) tagless protein in a single purification step with high clearance of host cell DNA, tagged precursor, higher and lower molecular weight impurities. Based on these preliminary results, we propose the iCapTag™ as a universal capture platform for diverse classes of recombinant therapeutic proteins.
Collapse
Affiliation(s)
- Sai Vivek Prabhala
- William G. Lowrie Department of Chemical and Biomolecular Engineering, Ohio State University, Columbus, OH 43210, United States
| | - Brian Marshall
- Eli Lilly and Company, Indianapolis, IN 46285, United States
| | | | - Yamin Fan
- Johnson & Johnson, 4560 Jinke Road, Shanghai 201210, China
| | - Ekaterina Creamer
- Ohio State Biochemistry Program, Ohio State University, Columbus, OH 43210, United States
| | - David W Wood
- William G. Lowrie Department of Chemical and Biomolecular Engineering, Ohio State University, Columbus, OH 43210, United States; Protein Capture Science LLC, Columbus, OH 43212, United States.
| |
Collapse
|
7
|
Roman-Ramos H, Ho PL. Current Technologies in Snake Venom Analysis and Applications. Toxins (Basel) 2024; 16:458. [PMID: 39591213 PMCID: PMC11598588 DOI: 10.3390/toxins16110458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
This comprehensive review explores the cutting-edge advancements in snake venom research, focusing on the integration of proteomics, genomics, transcriptomics, and bioinformatics. Highlighting the transformative impact of these technologies, the review delves into the genetic and ecological factors driving venom evolution, the complex molecular composition of venoms, and the regulatory mechanisms underlying toxin production. The application of synthetic biology and multi-omics approaches, collectively known as venomics, has revolutionized the field, providing deeper insights into venom function and its therapeutic potential. Despite significant progress, challenges such as the functional characterization of toxins and the development of cost-effective antivenoms remain. This review also discusses the future directions of venom research, emphasizing the need for interdisciplinary collaborations and new technologies (mRNAs, cryo-electron microscopy for structural determinations of toxin complexes, synthetic biology, and other technologies) to fully harness the biomedical potential of venoms and toxins from snakes and other animals.
Collapse
Affiliation(s)
- Henrique Roman-Ramos
- Laboratório de Biotecnologia, Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo 01504-001, SP, Brazil;
| | - Paulo Lee Ho
- Centro Bioindustrial, Instituto Butantan, São Paulo 05503-900, SP, Brazil
| |
Collapse
|
8
|
İncir İ, Kaplan Ö. Escherichia coli in the production of biopharmaceuticals. Biotechnol Appl Biochem 2024. [PMID: 39245907 DOI: 10.1002/bab.2664] [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: 04/23/2024] [Accepted: 08/23/2024] [Indexed: 09/10/2024]
Abstract
Escherichia coli has shouldered a massive workload with the discovery of recombinant DNA technology. A new era began in the biopharmaceutical industry with the production of insulin, the first recombinant protein, in E. coli and its use in treating diabetes. After insulin, many biopharmaceuticals produced from E. coli have been approved by the US Food and Drug Administration and the European Medicines Agency to treat various human diseases. Although E. coli has some disadvantages, such as lack of post-translational modifications and toxicity, it is an important host with advantages such as being a well-known bacterium in recombinant protein production, cheap, simple production system, and high yield. This study examined biopharmaceuticals produced and approved in E. coli under the headings of peptides, hormones, enzymes, fusion proteins, antibody fragments, vaccines, and other pharmaceuticals. The topics on which these biopharmaceuticals were approved for treating human diseases, when and by which company they were produced, and their use and development in the field are included.
Collapse
Affiliation(s)
- İbrahim İncir
- Kazım Karabekir Vocational School, Department of Medical Services and Techniques, Environmental Health Program, Karamanoğlu Mehmetbey University, Karaman, Turkey
| | - Özlem Kaplan
- Rafet Kayış Faculty of Engineering, Department of Genetics and Bioengineering, Alanya Alaaddin Keykubat University, Antalya, Turkey
| |
Collapse
|
9
|
Dannemeyer M, Berling A, Kanje S, Enstedt H, Xu L, Afshari D, Westin M, Hober G, Uhlén M, Hober S, Tegel H. Fast and robust recombinant protein production utilizing episomal stable pools in WAVE bioreactors. Protein Expr Purif 2024; 221:106505. [PMID: 38768672 DOI: 10.1016/j.pep.2024.106505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Protein reagents are essential resources for several stages of drug discovery projects from structural biology and assay development through lead optimization. Depending on the aim of the project different amounts of pure protein are required. Small-scale expressions are initially used to determine the reachable levels of production and quality before scaling up protein reagent supply. Commonly, amounts of several hundreds of milligrams to grams are needed for different experiments, including structural investigations and activity evaluations, which require rather large cultivation volumes. This implies that cultivation of large volumes of either transiently transfected cells or stable pools/stable cell lines is needed. Hence, a production process that is scalable, speeds up the development projects, and increases the robustness of protein reagent quality throughout scales. Here we present a protein production pipeline with high scalability. We show that our protocols for protein production in Chinese hamster ovary cells allow for a seamless and efficient scale-up with robust product quality and high performance. The flexible scale of the production process, as shown here, allows for shorter lead times in drug discovery projects where there is a reagent demand for a specific protein or a set of target proteins.
Collapse
Affiliation(s)
- Melanie Dannemeyer
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - Anna Berling
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - Sara Kanje
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - Henric Enstedt
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - LanLan Xu
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - Delaram Afshari
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - Malin Westin
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - Gabriella Hober
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - Mathias Uhlén
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - Sophia Hober
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden
| | - Hanna Tegel
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-10691, Stockholm, Sweden.
| |
Collapse
|
10
|
Chen H, Zhu J, Niu X, Cheng Y, Jian W, Gao F, Sunkang Y, Qi W, Huang L. Development of a P30 protein-based indirect ELISA for detecting African swine fever antibodies utilizing the HEK293F expression system. Vet J 2024; 306:106186. [PMID: 38936461 DOI: 10.1016/j.tvjl.2024.106186] [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: 04/27/2024] [Revised: 06/08/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
African swine fever (ASF) is an acute, febrile, and highly lethal infectious disease in pigs caused by the African swine fever virus (ASFV). Effective detection methods and strict biosecurity measures are crucial for preventing and controlling ASF, especially since there are currently no commercially available vaccines or antiviral drugs to combat ASFV infection effectively. However, the emergence of low-virulence strains of ASFV in recent years has led to false-positive results, highlighting the importance of early-produced antibody detection methods. Therefore, detecting antibodies against ASFV produced early in the infection can facilitate the prompt identification of infected pigs. This study focused on the p30 protein, an early expressed protein during ASFV infection, to develop an indirect ELISA. This method was established using the HEK293F suspension cell expression system, which has the ability to produce large quantities of correctly folded proteins with normal functionality. In this study, we developed an indirect ELISA test utilizing the p30 recombinant protein produced by the HEK293F suspension cell expression system as the antigen coating. The concentration of the p30 protein obtained from the HEK293F suspension cell expression system was measured at 4.668 mg/mL, serving as the foundation for establishing the indirect ELISA. Our findings indicate that the indirect ELISA method exhibits a sensitivity of 1:12800. Furthermore, it demonstrates high specificity and excellent reproducibility. Comparing our results to those obtained from the commercial kit, we found a coincidence rate of 98.148 % for the indirect ELISA. In summary, we have developed a sensitive method for detecting ASFV, providing a valuable tool for monitoring ASFV infection in pig herds.
Collapse
Affiliation(s)
- Huahan Chen
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China
| | - Junhai Zhu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China
| | - Xuefeng Niu
- State Key Laboratory of Respiratory Disease, Guangzhou 510120, China; National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China; National Center for Respiratory Medicine, Guangzhou 510120, China; Guangzhou Institute of Respiratory Health, Guangzhou 510120, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yuanyi Cheng
- State Key Laboratory of Respiratory Disease, Guangzhou 510120, China; National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China; National Center for Respiratory Medicine, Guangzhou 510120, China; Guangzhou Institute of Respiratory Health, Guangzhou 510120, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Weijun Jian
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China
| | - Fei Gao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China
| | - Yongjie Sunkang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China
| | - Wenbao Qi
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China.
| | - Lihong Huang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou 510642, China.
| |
Collapse
|
11
|
Zong HF, Li X, Han L, Wang L, Liu JJ, Yue YL, Chen J, Ke Y, Jiang H, Xie YQ, Zhang BH, Zhu JW. A novel bispecific antibody drug conjugate targeting HER2 and HER3 with potent therapeutic efficacy against breast cancer. Acta Pharmacol Sin 2024; 45:1727-1739. [PMID: 38605180 PMCID: PMC11272928 DOI: 10.1038/s41401-024-01279-8] [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: 11/16/2023] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Antibody drug conjugate (ADC) therapy has become one of the most promising approaches in cancer immunotherapy. Bispecific targeting could enhance the efficacy and safety of ADC by improving its specificity, affinity and internalization. In this study we constructed a HER2/HER3-targeting bispecific ADC (BsADC) and characterized its physiochemical properties, target specificity and internalization in vitro, and assessed its anti-tumor activities in breast cancer cell lines and in animal models. The HER2/HER3-targeting BsADC had a drug to antibody ratio (DAR) of 2.89, displayed a high selectivity against the target JIMT-1 breast cancer cells in vitro, as well as a slightly higher level of internalization than HER2- or HER3-monospecific ADCs. More importantly, the bispecific ADC potently inhibited the viability of MCF7, JIMT-1, BT474, BxPC-3 and SKOV-3 cancer cells in vitro. In JIMT-1 breast cancer xenograft mice, a single injection of bispecific ADC (3 mg/kg, i.v.) significantly inhibited the tumor growth with an efficacy comparable to that caused by combined injection of HER2 and HER3-monospecific ADCs (3 mg/kg for each). Our study demonstrates that the bispecific ADC concept can be applied to development of more potent new cancer therapeutics than the monospecific ADCs.
Collapse
Affiliation(s)
- Hui-Fang Zong
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Jecho Institute Co., Ltd., Shanghai, 200240, China
| | - Xi Li
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lei Han
- Jecho Institute Co., Ltd., Shanghai, 200240, China
| | - Lei Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jun-Jun Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ya-Li Yue
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jie Chen
- Jecho Institute Co., Ltd., Shanghai, 200240, China
| | - Yong Ke
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hua Jiang
- Jecho Laboratories, Inc., Frederick, MD, 21704, USA
| | - Yue-Qing Xie
- Jecho Institute Co., Ltd., Shanghai, 200240, China
- Jecho Laboratories, Inc., Frederick, MD, 21704, USA
| | - Bao-Hong Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian-Wei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Jecho Institute Co., Ltd., Shanghai, 200240, China.
- Jecho Laboratories, Inc., Frederick, MD, 21704, USA.
| |
Collapse
|
12
|
Masuda K, Kubota M, Nakazawa Y, Iwama C, Watanabe K, Ishikawa N, Tanabe Y, Kono S, Tanemura H, Takahashi S, Makino T, Okumura T, Horiuchi T, Nonaka K, Murakami S, Kamihira M, Omasa T. Establishment of a novel cell line, CHO-MK, derived from Chinese hamster ovary tissues for biologics manufacturing. J Biosci Bioeng 2024; 137:471-479. [PMID: 38472071 DOI: 10.1016/j.jbiosc.2024.02.005] [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: 12/07/2023] [Revised: 02/04/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024]
Abstract
Chinese hamster ovary (CHO) cells are widely used as a host for producing recombinant therapeutic proteins due to advantages such as human-like post-translational modification, correct protein folding, higher productivity, and a proven track record in biopharmaceutical development. Much effort has been made to improve the process of recombinant protein production, in terms of its yield and productivity, using conventional CHO cell lines. However, to the best of our knowledge, no attempts have been made to acquire new CHO cell lines from Chinese hamster ovary. In this study, we established and characterized a novel CHO cell line, named CHO-MK, derived from freshly isolated Chinese hamster ovary tissues. Some immortalized cell lines were established via sub-culture derived from primary culture, one of which was selected for further development toward a unique expression system design. After adapting serum-free and suspension culture conditions, the resulting cell line exhibited a considerably shorter doubling time (approximately 10 h) than conventional CHO cell lines (approximately 20 h). Model monoclonal antibody (IgG1)-producing cells were generated, and the IgG1 concentration of fed-batch culture reached approximately 5 g/L on day 8 in a 200-L bioreactor. The cell bank of CHO-MK cells was prepared as a new host and assessed for contamination by adventitious agents, with the results indicating that it was free from any such contaminants, including infectious viruses. Taking these findings together, this study showed the potential of CHO-MK cells with a shorter doubling time/process time and enhanced productivity in biologics manufacturing.
Collapse
Affiliation(s)
- Kenji Masuda
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan; Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Michi Kubota
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Chitose Laboratory Corp., KSP EAST511, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Yuto Nakazawa
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Chigusa Iwama
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Chitose Laboratory Corp., KSP EAST511, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Kazuhiko Watanabe
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Naoto Ishikawa
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Yumiko Tanabe
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Satoru Kono
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Hiroki Tanemura
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Shinichi Takahashi
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Tomohiro Makino
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan
| | - Takeshi Okumura
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan; Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Takayuki Horiuchi
- Chitose Laboratory Corp., KSP EAST511, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Koichi Nonaka
- Biologics Division, Biologics Technology Research Laboratories I, Daiichi Sankyo Co., Ltd., 2716-1 Kurakake, Akaiwa, Chiyoda-machi, Gunma 370-0503, Japan; Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Sei Murakami
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan
| | - Masamichi Kamihira
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takeshi Omasa
- Manufacturing Technology Association of Biologics, 2-6-16 Shinkawa, Chuo-ku, Tokyo 104-0033, Japan; Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
13
|
Panikulam S, Hanke A, Kroener F, Karle A, Anderka O, Villiger TK, Lebesgue N. Host cell protein networks as a novel co-elution mechanism during protein A chromatography. Biotechnol Bioeng 2024; 121:1716-1728. [PMID: 38454640 DOI: 10.1002/bit.28678] [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: 11/03/2023] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 03/09/2024]
Abstract
Host cell proteins (HCPs) are process-related impurities of therapeutic proteins produced in for example, Chinese hamster ovary (CHO) cells. Protein A affinity chromatography is the initial capture step to purify monoclonal antibodies or Fc-based proteins and is most effective for HCP removal. Previously proposed mechanisms that contribute to co-purification of HCPs with the therapeutic protein are either HCP-drug association or leaching from chromatin heteroaggregates. In this study, we analyzed protein A eluates of 23 Fc-based proteins by LC-MS/MS to determine their HCP content. The analysis revealed a high degree of heterogeneity in the number of HCPs identified in the different protein A eluates. Among all identified HCPs, the majority co-eluted with less than three Fc-based proteins indicating a drug-specific co-purification for most HCPs. Only ten HCPs co-purified with over 50% of the 23 Fc-based proteins. A correlation analysis of HCPs identified across multiple protein A eluates revealed their co-elution as HCP groups. Functional annotation and protein interaction analysis confirmed that some HCP groups are associated with protein-protein interaction networks. Here, we propose an additional mechanism for HCP co-elution involving protein-protein interactions within functional networks. Our findings may help to guide cell line development and to refine downstream purification strategies.
Collapse
Affiliation(s)
- Sherin Panikulam
- Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Alexander Hanke
- Analytical Development and Characterization, Biopharmaceutical Product and Process Development, Technical Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - Frieder Kroener
- Analytical Development and Characterization, Biopharmaceutical Product and Process Development, Technical Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - Anette Karle
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Oliver Anderka
- Analytical Development and Characterization, Biopharmaceutical Product and Process Development, Technical Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - Thomas K Villiger
- Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland
| | - Nicolas Lebesgue
- Analytical Development and Characterization, Biopharmaceutical Product and Process Development, Technical Research and Development, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
14
|
Kumar V, Barwal A, Sharma N, Mir DS, Kumar P, Kumar V. Therapeutic proteins: developments, progress, challenges, and future perspectives. 3 Biotech 2024; 14:112. [PMID: 38510462 PMCID: PMC10948735 DOI: 10.1007/s13205-024-03958-z] [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: 06/03/2023] [Accepted: 02/13/2024] [Indexed: 03/22/2024] Open
Abstract
Proteins are considered magic molecules due to their enormous applications in the health sector. Over the past few decades, therapeutic proteins have emerged as a promising treatment option for various diseases, particularly cancer, cardiovascular disease, diabetes, and others. The formulation of protein-based therapies is a major area of research, however, a few factors still hinder the large-scale production of these therapeutic products, such as stability, heterogenicity, immunogenicity, high cost of production, etc. This review provides comprehensive information on various sources and production of therapeutic proteins. The review also summarizes the challenges currently faced by scientists while developing protein-based therapeutics, along with possible solutions. It can be concluded that these proteins can be used in combination with small molecular drugs to give synergistic benefits in the future.
Collapse
Affiliation(s)
- Vimal Kumar
- University Institute of Biotechnology, Chandigarh University, Gharuan, Mohali, Punjab 140413 India
| | - Arti Barwal
- Department of Microbial Biotechnology, Panjab University, South Campus, Sector-25, Chandigarh, 160014 India
| | - Nitin Sharma
- Department of Biotechnology, Chandigarh Group of Colleges, Mohali, Punjab 140307 India
| | - Danish Shafi Mir
- University Institute of Biotechnology, Chandigarh University, Gharuan, Mohali, Punjab 140413 India
| | - Pradeep Kumar
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, 173229 India
| | - Vikas Kumar
- University Institute of Biotechnology, Chandigarh University, Gharuan, Mohali, Punjab 140413 India
| |
Collapse
|
15
|
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
|
16
|
Clarke EC. Considerations for Glycoprotein Production. Methods Mol Biol 2024; 2762:329-351. [PMID: 38315375 DOI: 10.1007/978-1-0716-3666-4_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
This chapter is intended to provide insights for researchers aiming to choose an appropriate expression system for the production of recombinant glycoproteins. Producing glycoproteins is complex, as glycosylation patterns are determined by the availability and abundance of specific enzymes rather than a direct genetic blueprint. Furthermore, the cell systems often employed for protein production are evolutionarily distinct, leading to significantly different glycosylation when utilized for glycoprotein production. The selection of an appropriate production system depends on the intended applications and desired characteristics of the protein. Whether the goal is to produce glycoproteins mimicking native conditions or to intentionally alter glycan structures for specific purposes, such as enhancing immunogenicity in vaccines, understanding glycosylation present in the different systems and in different growth conditions is essential. This chapter will cover Escherichia coli, baculovirus/insect cell systems, Pichia pastoris, as well as different mammalian cell culture systems including Chinese hamster ovary (CHO) cells, human endothelial kidney (HEK) cell lines, and baby hamster kidney (BHK) cells.
Collapse
Affiliation(s)
- Elizabeth C Clarke
- Center for Global Health, Division of Infectious Diseases, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
17
|
Vitharana S, Stillahn JM, Katayama DS, Henry CS, Manning MC. Application of Formulation Principles to Stability Issues Encountered During Processing, Manufacturing, and Storage of Drug Substance and Drug Product Protein Therapeutics. J Pharm Sci 2023; 112:2724-2751. [PMID: 37572779 DOI: 10.1016/j.xphs.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
The field of formulation and stabilization of protein therapeutics has become rather extensive. However, most of the focus has been on stabilization of the final drug product. Yet, proteins experience stress and degradation through the manufacturing process, starting with fermentaition. This review describes how formulation principles can be applied to stabilize biopharmaceutical proteins during bioprocessing and manufacturing, considering each unit operation involved in prepration of the drug substance. In addition, the impact of the container on stabilty is discussed as well.
Collapse
Affiliation(s)
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
18
|
Yoon C, Baek KE, Kim D, Lee GM. Mitigating transcriptional bottleneck using a constitutively active transcription factor, VP16-CREB, in mammalian cells. Metab Eng 2023; 80:33-44. [PMID: 37709006 DOI: 10.1016/j.ymben.2023.09.005] [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: 08/13/2022] [Revised: 07/13/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
High-level expression of recombinant proteins in mammalian cells has long been an area of interest. Inefficient transcription machinery is often an obstacle in achieving high-level expression of recombinant proteins in mammalian cells. Synthetic promoters have been developed to improve the transcription efficiency, but have achieved limited success due to the limited availability of transcription factors (TFs). Here, we present a TF-engineering approach to mitigate the transcriptional bottlenecks of recombinant proteins. This includes: (i) identification of cAMP response element binding protein (CREB) as a candidate TF by searching for TFs enriched in the cytomegalovirus (CMV) promoter-driven high-producing recombinant Chinese hamster ovary (rCHO) cell lines via transcriptome analysis, (ii) confirmation of transcriptional limitation of active CREB in rCHO cell lines, and (iii) direct activation of the transgene promoter by expressing constitutively active CREB at non-cytotoxic levels in rCHO cell lines. With the expression of constitutively active VP16-CREB, the production of therapeutic proteins, such as monoclonal antibody and etanercept, in CMV promoter-driven rCHO cell lines was increased up to 3.9-fold. VP16-CREB was also used successfully with synthetic promoters containing cAMP response elements. Taken together, this strategy to introduce constitutively active TFs into cells is a useful means of overcoming the transcriptional limitations in recombinant mammalian cells.
Collapse
Affiliation(s)
- Chansik Yoon
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Kyoung Eun Baek
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Dongil Kim
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
19
|
Kim Y, Jahan UM, Deltchev AP, Lavrik N, Reukov V, Minko S. Strategy for Nonenzymatic Harvesting of Cells via Decoupling of Adhesive and Disjoining Domains of Nanostructured Stimulus-Responsive Polymer Films. ACS APPLIED MATERIALS & INTERFACES 2023; 15:49012-49021. [PMID: 37824473 PMCID: PMC10614186 DOI: 10.1021/acsami.3c11296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/28/2023] [Indexed: 10/14/2023]
Abstract
The nanostructured polymer film introduces a novel mechanism of nonenzymatic cell harvesting by decoupling solid cell-adhesive and soft stimulus-responsive cell-disjoining areas on the surface. The key characteristics of this architecture are the decoupling of adhesion from detachment and the impermeability to the integrin protein complex of the adhesive domains. This surface design eliminates inherent limitations of thermoresponsive coatings, namely, the necessity for the precise thickness of the coating, grafting or cross-linking density, and material of the basal substrate. The concept is demonstrated with nanostructured thermoresponsive films made of cell-adhesive epoxy photoresist domains and cell-disjoining poly(N-isopropylacrylamide) brush domains.
Collapse
Affiliation(s)
- Yongwook Kim
- Nanostructured
Material Lab, University of Georgia, Athens, Georgia 30602, United States
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
- Lawrence
Livermore National Lab, Livermore, California 94500, United States
| | - Ummay Mowshome Jahan
- Department
of Textiles, Merchandising, and Interiors, University of Georgia, Athens, Georgia 30602, United States
- Department
of Textile Engineering, Chemistry and Science, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Alexander Pennef Deltchev
- Nanostructured
Material Lab, University of Georgia, Athens, Georgia 30602, United States
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Nickolay Lavrik
- Center
for Nanophase Materials Sciences, Oak Ridge
National Lab, Oak Ridge, Tennessee 37831, United States
| | - Vladimir Reukov
- Department
of Textiles, Merchandising, and Interiors, University of Georgia, Athens, Georgia 30602, United States
| | - Sergiy Minko
- Nanostructured
Material Lab, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
20
|
Maltais JS, Lord-Dufour S, Morasse A, Stuible M, Loignon M, Durocher Y. Repressing expression of difficult-to-express recombinant proteins during the selection process increases productivity of CHO stable pools. Biotechnol Bioeng 2023; 120:2840-2852. [PMID: 37232536 DOI: 10.1002/bit.28435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023]
Abstract
More than half of licensed therapeutic recombinant proteins (r-proteins) are manufactured using constitutively-expressing, stably-transfected Chinese hamster ovary (CHO) clones. While constitutive CHO expression systems have proven their efficacy for the manufacturing of monoclonal antibodies, many next-generation therapeutics such as cytokines and bispecific antibodies as well as biological targets such as ectodomains of transmembrane receptors remain intrinsically challenging to produce. Herein, we exploited a cumate-inducible CHO platform allowing reduced expression of various classes of r-proteins during selection of stable pools. Following stable pool generation, fed-batch productions showed that pools generated without cumate (OFF-pools) were significantly more productive than pools selected in the presence of cumate (ON-pools) for 8 out of the 10 r-proteins tested, including cytokines, G-protein coupled receptors (GPCRs), the HVEM membrane receptor ectodomain, the multifunctional protein High Mobility Group protein B1 (HMGB1), as well as monoclonal and bispecific T-cell engager antibodies. We showed that OFF-pools contain a significantly larger proportion of cells producing high levels of r-proteins and that these cells tend to proliferate faster when expression is turned off, suggesting that r-protein overexpression imposes a metabolic burden on the cells. Cell viability was lower and pool recovery was delayed during selection of ON-pools (mimicking constitutive expression), suggesting that high producers were likely lost or overgrown by faster-growing, low-producing cells. We also observed a correlation between the expression levels of the GPCRs with Binding immunoglobulin Protein, an endoplasmic reticulum (ER) stress marker. Taken together, these data suggest that using an inducible system to minimize r-protein expression during stable CHO pool selection reduces cellular stresses, including ER stress and metabolic burden, leading to pools with greater frequency of high-expressing cells, resulting in improved volumetric productivity.
Collapse
Affiliation(s)
- Jean-Sébastien Maltais
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Simon Lord-Dufour
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Audrey Morasse
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Matthew Stuible
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Martin Loignon
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Yves Durocher
- Mammalian Cell Expression, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
21
|
Ben Yahia B, Piednoir A, Dahomais T, Eggermont S, Paul W. "Organized stress" for robust scale-up of intensified production process with fed-batch seed bioreactor. Biotechnol Bioeng 2023; 120:2509-2522. [PMID: 37027375 DOI: 10.1002/bit.28396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/08/2023]
Abstract
Process intensification has been widely used for many years in the mammalian biomanufacturing industry to increase productivity, agility and flexibility while reducing production costs. The most commonly used intensified processes are operated using a perfusion or fed-batch seed bioreactor enabling a higher than usual seeding density in the fed-batch production bioreactor. Hence, as part of the growth phase is shifted to the seed bioreactor, there is a lower split ratio, which increases the criticality of the seed bioreactor and could impact production performance. Therefore, such intensified processes should be designed and characterized for robust process scale-up. This research work is focused on intensified processes with high seeding density inoculated from seed bioreactor in fed-batch mode. The impact of the feeding strategy and specific power input (P/V) in the seed bioreactor and on the production step with two different cell lines (CL1 and CL2) producing two different monoclonal antibodies was investigated. Cell culture performance in the production bioreactor has been improved due to more stressful conditions for the cells in the seed bioreactor and the impact of the production bioreactor P/V on the production performance was limited. This is the first reported study highlighting a positive impact of cellular stress in seed bioreactors on intensified production bioreactor with the introduction of the "organized stress" concept.
Collapse
Affiliation(s)
- Bassem Ben Yahia
- Biologics Process Sciences, Biotech Sciences, UCB Pharma S.A., Avenue de l'Industrie, Brussels, Braine l'Alleud, Belgium
| | - Antoine Piednoir
- Biologics Process Sciences, Biotech Sciences, UCB Pharma S.A., Avenue de l'Industrie, Brussels, Braine l'Alleud, Belgium
| | - Thomas Dahomais
- Biologics Process Sciences, Biotech Sciences, UCB Pharma S.A., Avenue de l'Industrie, Brussels, Braine l'Alleud, Belgium
| | - Stefanie Eggermont
- Biologics Process Sciences, Biotech Sciences, UCB Pharma S.A., Avenue de l'Industrie, Brussels, Braine l'Alleud, Belgium
| | - Wolfgang Paul
- Biologics Process Sciences, Biotech Sciences, UCB Pharma S.A., Avenue de l'Industrie, Brussels, Braine l'Alleud, Belgium
| |
Collapse
|
22
|
Murashkin DE, Belenkaya SV, Bondar AA, Elchaninov VV, Shcherbakov DN. Analysis of Some Biochemical Properties of Recombinant Siberian Roe Deer (Capreolus pygargus) Chymosin Obtained in the Mammalian Cell Culture (CHO-K1). BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1284-1295. [PMID: 37770395 DOI: 10.1134/s0006297923090080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 09/30/2023]
Abstract
Structure of the chymosin gene of Siberian roe deer (Capreolus pygargus) was established for the first time and its exon/intron organization was determined. Coding part of the chymosin gene of C. pygargus was reconstructed by the Golden Gate method and obtained as a DNA clone. Comparative sequence analysis of the roe deer, cow, and one-humped camel prochymosins revealed a number of amino acid substitutions at the sites forming the substrate-binding cavity of the enzyme and affecting the S4 and S1' + S3' specificity subsites. Integration vector pIP1 was used to construct a plasmid pIP1-Cap in order to express recombinant roe deer prochymosin gene in CHO-K1 cells. CHO-K1-CYM-Cap pool cells were obtained, allowing synthesis and secretion of recombinant prochymosin into the culture fluid. As a result of zymogen activation, a recombinant roe deer chymosin was obtained and its total milk-clotting activity was estimated to be 468.4 ± 11.1 IMCU/ml. Yield of the recombinant roe deer chymosin was 500 mg/liter or ≈468,000 IMCU/liter, which exceeds the yields of genetically engineered chymosins in most of the expression systems used. Basic biochemical properties of the obtained enzyme were compared with the commercial preparations of recombinant chymosins from one-humped camel (Camelus dromedarius) and cow (Bos taurus). Specific milk-clotting activity of the recombinant chymosin of C. pygargus was 938 ± 22 IMCU/mg, which was comparable to that of the reference enzymes. Non-specific proteolytic activity of the recombinant roe deer chymosin was 1.4-4.5 times higher than that of the cow and camel enzymes. In terms of coagulation specificity, recombinant chymosin of C. pygargus occupied an intermediate position between the genetically engineered analogs of B. taurus and C. dromedarius chymosins. Thermostability threshold of the recombinant roe deer chymosin was 55°C. At 60°C, the enzyme retained <1% of its initial milk-clotting activity, and its complete thermal inactivation was observed at 65°C.
Collapse
Affiliation(s)
- Denis E Murashkin
- State Research Center of Virology and Biotechnology VECTOR, Koltsovo, 630559, Russia
| | - Svetlana V Belenkaya
- State Research Center of Virology and Biotechnology VECTOR, Koltsovo, 630559, Russia.
- Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Aleksandr A Bondar
- Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Vadim V Elchaninov
- Federal Altaic Scientific Center of Agrobiotechnology, Barnaul, 656910, Russia
| | - Dmitrii N Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Koltsovo, 630559, Russia
- Altai State University, Barnaul, 656049, Russia
| |
Collapse
|
23
|
Sou SN, Harris CL, Williams R, Kozub D, Zurlo F, Patel YD, Kallamvalli Illam Sankaran P, Daramola O, Brown A, James DC, Hatton D, Dunn S, Gibson SJ. CHO synthetic promoters improve expression and product quality of biotherapeutic proteins. Biotechnol Prog 2023; 39:e3348. [PMID: 37114854 DOI: 10.1002/btpr.3348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/27/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023]
Abstract
When expressing complex biotherapeutic proteins, traditional expression plasmids and methods may not always yield sufficient levels of high-quality product. High-strength viral promoters commonly used for recombinant protein (rProtein) production in mammalian cells allow for maximal expression, but provide limited scope to alter their transcription dynamics. However, synthetic promoters designed to provide tunable transcriptional activity offer a plasmid engineering approach to more precisely regulate product quality, yield or to reduce product related contaminants. We substituted the viral promoter CMV with synthetic promoters that offer different transcriptional activities to express our gene of interest in Chinese hamster ovary (CHO) cells. Stable pools were established and the benefits of regulating transgene transcription on the quality of biotherapeutics were examined in stable pool fed-batch overgrow experiments. Specific control of gene expression of the heavy chain (HC):light chain (LC) of a Fab, and the ratio between the two HCs in a Duet mAb reduced levels of aberrant protein contaminants; and the controlled expression of the helper gene XBP-1s improved expression of a difficult-to-express mAb. This synthetic promoter technology benefits applications that require custom activity. Our work highlights the advantages of employing synthetic promoters for production of more complex rProteins.
Collapse
Affiliation(s)
- Si Nga Sou
- BioPharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Claire L Harris
- BioPharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Dorota Kozub
- BioPharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Fabio Zurlo
- BioPharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Yash D Patel
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | | | | | - Adam Brown
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - David C James
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Diane Hatton
- BioPharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | - Sarah Dunn
- BioPharmaceutical Development, R&D, AstraZeneca, Cambridge, UK
| | | |
Collapse
|
24
|
Zhang Y, Li SY, Zhu HJ, Lai JW, Sun SS, Lin Y, Li XL, Guo ZB, Lv Z, Meng H, Hu K, Xu M, Yu TT. Mechano-biomimetic hydrogel 3D cell cultivation as a strategy to improve mammalian cell protein expression. Mater Today Bio 2023; 21:100732. [PMID: 37521005 PMCID: PMC10371807 DOI: 10.1016/j.mtbio.2023.100732] [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: 03/31/2023] [Revised: 06/23/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023] Open
Abstract
Eukaryotic expression systems are frequently employed for the production of recombinant proteins as therapeutics as well as research tools. Among which mammalian cell protein expression approach is the most powerful one, which can express complex proteins or genetic engineered biological drugs, such as PD-1. However, the high expense, which partially derives from its low protein yielding efficiency, limited the further application of such approach in large scale production of target proteins. To address this issue, we proposed a novel technique to promote the protein production efficiency of mammal cells without using conventional genetic engineered approaches. By placing 293T cells in a hydrogel 3D cell culture platform and adjusting the stress relaxation of the matrix hydrogel, cells formed multicellular spheroids by self-organization. In particular, the multicellular spheroids have a significantly enhanced ability to transiently express multiple proteins (SHH-N, PD-1 and PDL-1). We also examined in detail the mechanism underlying this phenomenon, and found that the reorganization of cytoskeleton during spheroids formation enhances the translation process of protein by recruiting ribosomes. Overall, this finding provides a novel approach for subsequent improvement of large-scale mammalian protein expression cell systems.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Si-yang Li
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Hang-ju Zhu
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Jun-Wei Lai
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Shuo-shuo Sun
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Yue Lin
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Xing-ling Li
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Zhao-bin Guo
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ziheng Lv
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Hongxu Meng
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 314400, China
| | - Ke Hu
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Ming Xu
- Engineering Research Center of Health Emergency, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
- Jiangsu Province Engineering Research Center of Health Emergency, Nanjing, 210009, China
- Jiangsu Preventive Medicine Association, Nanjing, 210009, China
- School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ting-ting Yu
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Preventive Medicine Association, Nanjing, 210009, China
- School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
25
|
Wang Y, Zhang K, Zhao Y, Li Y, Su W, Li S. Construction and Applications of Mammalian Cell-Based DNA-Encoded Peptide/Protein Libraries. ACS Synth Biol 2023; 12:1874-1888. [PMID: 37315219 DOI: 10.1021/acssynbio.3c00043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
DNA-encoded peptide/protein libraries are the starting point for protein evolutionary modification and functional peptide/antibody selection. Different display technologies, protein directed evolution, and deep mutational scanning (DMS) experiments employ DNA-encoded libraries to provide sequence variations for downstream affinity- or function-based selections. Mammalian cells promise the inherent post-translational modification and near-to-natural conformation of exogenously expressed mammalian proteins and thus are the best platform for studying transmembrane proteins or human disease-related proteins. However, due to the current technical bottlenecks of constructing mammalian cell-based large size DNA-encoded libraries, the advantages of mammalian cells as screening platforms have not been fully exploited. In this review, we summarize the current efforts in constructing DNA-encoded libraries in mammalian cells and the existing applications of these libraries in different fields.
Collapse
Affiliation(s)
- Yi Wang
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Kaili Zhang
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yanjie Zhao
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yifan Li
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Weijun Su
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuai Li
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
26
|
Chen J, Pan Z, Han L, Liu J, Yue Y, Xiao X, Zhang B, Wu M, Yuan Y, Bian Y, Jiang H, Xie Y, Zhu J. Binding domain on CD22 molecules contributing to the biological activity of T cell-engaging bispecific antibodies. Heliyon 2023; 9:e17960. [PMID: 37456045 PMCID: PMC10344817 DOI: 10.1016/j.heliyon.2023.e17960] [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: 12/31/2022] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023] Open
Abstract
CD22, as the B-cell malignancies antigen, has been targeted for immunotherapies through CAR-T cells, antibody-drug conjugates (ADCs) and immunotoxins via interaction of antibodies with binding domains on the receptor. We hypothesized that avidity and binding domain of antibody to target cells may have significant impact on the biological function in tumor immunotherapy, and T cell-engaging bispecific antibody (TCB) targeting CD22 could be used in the therapy of hematologic malignancies. So, to address the question, we utilized the information of six previously reported CD22 mAbs to generate CD22-TCBs with different avidity to different domains on CD22 protein. We found that the avidity of CD22-TCBs to protein was not consistent with the avidity to target cells, indicating that TCBs had different binding mode to the protein and cells. In vitro results indicated that CD22-TCBs mediated cytotoxicity depended on the avidity of antibodies to target cells rather than to protein. Moreover, distal binding domain of the antigen contributed to the avidity and biological activity of IgG-[L]-scfv-like CD22-TCBs. The T cells' proliferation, activation, cytotoxicity as well as cytokine release were compared, and G5/44 BsAb was selected for further in vivo assessment in anti-tumor activity. In vivo results demonstrated that CD22-TCB (G5/44 BsAb) significantly inhibited the tumors growth in mice. All these data suggested that CD22-TCBs could be developed as a promising candidate for B-cell malignancies therapy through optimizing the design with avidity and binding domain to CD22 target in consideration.
Collapse
Affiliation(s)
- Jie Chen
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhidi Pan
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lei Han
- Jecho Institute, Shanghai 200240, China
| | - Junjun Liu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yali Yue
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | | | - Baohong Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Mingyuan Wu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yunsheng Yuan
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yanlin Bian
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hua Jiang
- Jecho Biopharmaceuticals Co., Ltd, Tianjin, 300450, China
- Jecho Laboratories, Inc., Frederick, MD21704, USA
| | - Yueqing Xie
- Jecho Laboratories, Inc., Frederick, MD21704, USA
| | - Jianwei Zhu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
27
|
Srila W, Baumann M, Riedl M, Rangnoi K, Borth N, Yamabhai M. Glutamine synthetase (GS) knockout (KO) using CRISPR/Cpf1 diversely enhances selection efficiency of CHO cells expressing therapeutic antibodies. Sci Rep 2023; 13:10473. [PMID: 37380701 DOI: 10.1038/s41598-023-37288-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 06/19/2023] [Indexed: 06/30/2023] Open
Abstract
The glutamine synthetase (GS)-based Chinese hamster ovary (CHO) selection system is an attractive approach to efficiently identify suitable clones in the cell line generation process for biologics manufacture, for which GS-knockout (GS-KO) CHO cell lines are commonly used. Since genome analysis indicated that there are two GS genes in CHO cells, deleting only 1 GS gene could potentially result in the activation of other GS genes, consequently reducing the selection efficiency. Therefore, in this study, both GS genes identified on chromosome 5 (GS5) and 1 (GS1) of CHO-S and CHO-K1, were deleted using CRISPR/Cpf1. Both single and double GS-KO CHO-S and K1 showed robust glutamine-dependent growth. Next, the engineered CHO cells were tested for their efficiency of selection of stable producers of two therapeutic antibodies. Analysis of pool cultures and subclones after a single round of 25 µM methionine sulfoxinime (MSX) selection indicated that for CHO-K1 the double GS5,1-KO was more efficient as in the case of a single GS5-KO the GS1 gene was upregulated. In CHO-S, on the other hand, with an autologously lower level of expression of both variants of GS, a single GS5-KO was more robust and already enabled selection of high producers. In conclusion, CRISPR/Cpf1 can be efficiently used to knock out GS genes from CHO cells. The study also indicates that for the generation of host cell lines for efficient selection, the initial characterisation of expression levels of the target gene as well as the identification of potential escape mechanisms is important.
Collapse
Affiliation(s)
- Witsanu Srila
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Martina Baumann
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, Austria
| | - Markus Riedl
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, Austria
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Kuntalee Rangnoi
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Nicole Borth
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, Austria.
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria.
| | - Montarop Yamabhai
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand.
| |
Collapse
|
28
|
Lee H, Song ES, Lee YH, Park JY, Kuk MU, Kwon HW, Roh H, Park JT. A novel hybrid promoter capable of continuously producing proteins in high yield. Biochem Biophys Res Commun 2023; 650:103-108. [PMID: 36774687 DOI: 10.1016/j.bbrc.2023.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The establishment of cell lines with a high protein production is the most crucial objective in the field of biopharmaceuticals. To this end, efforts have been made to increase transgene expression through promoter improvement, but the efficiency or stability of protein production was insufficient for use in commercial production. Here, we developed a novel strategy to increase the efficiency and stability of protein production by hybridizing a promoter that exhibits higher expression levels at the transient level with a promoter that exhibits higher stability at the stable level. Expression levels of transgenes by each promoter were measured at transient and stable levels for five single promoters: Rous sarcoma virus (RSV), cytomegalovirus (CMV), human phosphoglycerate kinase (hPGK), simian virus 40 (SV40), and zebrafish ubiquitin B (Ubb). The hPGK promoter enabled high-yield transgene expression at transient levels and the SV40 promoter enabled sustained expression at stable levels. Therefore, hPGK and SV40 promoters were selected as candidates for establishing hybrid promoters and two hybrid promoters were constructed; one hybrid promoter in which the SV40 promoter is added before the hPGK promoter (a.k.a. SKYI) and the other hybrid promoter in which the SV40 promoter is added after the hPGK promoter (a.k.a. SKYII). Of the two hybrid promoters, the hybrid promoter SKYII promoted high-yield transgene expression at both transient and stable levels compared to single hPGK and SV40. Together, our findings open new doors in the field of biopharmaceuticals by presenting a novel promoter platform that can be used for high-yield and sustained protein production.
Collapse
Affiliation(s)
- Haneur Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Eun Seon Song
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Yun Haeng Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Ji Yun Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Myeong Uk Kuk
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Hyung Wook Kwon
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea; Convergence Research Center for Insect Vectors, Incheon National University, Incheon, 22012, South Korea
| | - Hyungmin Roh
- Department of Chemical and Biological Engineering, Inha Technical College, Incheon, 22212, South Korea.
| | - Joon Tae Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea; Convergence Research Center for Insect Vectors, Incheon National University, Incheon, 22012, South Korea.
| |
Collapse
|
29
|
Mullins DR, Galosy SS. Development of a novel capillary electrophoresis method for quantitative measurements of intracellular recombinant protein titer. J Biotechnol 2023; 365:54-61. [PMID: 36780953 DOI: 10.1016/j.jbiotec.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/12/2023] [Accepted: 02/09/2023] [Indexed: 02/13/2023]
Abstract
The measurement of volumetric titer is an integral step in the assessment and selection of a production cell line and cell culture process. The production of monoclonal antibodies (mAbs), a major class of therapeutic proteins, in Chinese Hamster Ovary (CHO) cell lines is challenging due to the clone-to-clone variations in the intrinsic capability to secrete a biologically complex protein. The measurement of intracellular mAb concentration could be a valuable tool to determine the ratio of intracellular to secreted product and be part of the evaluation of potential mAb productive cell lines. High throughput automation is a valuable tool that is used in bioprocess development to reduce work intensive steps. When coupled with the Simple Western (Wes) platform, automated capillary electrophoresis is an efficient method to measure recombinant protein concentration. In this study, we demonstrate the utility of using the automated Wes to rapidly measure intracellular titer and then compare the intracellular titer, volumetric titer and specific productivity between high and low production CHO clones expressing a model human IgG1 mAb.
Collapse
Affiliation(s)
- David R Mullins
- Global Investigative Safety, GlaxoSmithKline (GSK), Collegeville, PA, USA.
| | - Sybille S Galosy
- Biopharm Microbial & Cell Culture Development, GlaxoSmithKline (GSK), King of Prussia, PA, USA
| |
Collapse
|
30
|
Chen C, Wang Z, Sun Z, Li W, Dimitrov DS. Development of an efficient method for selection of stable cell pools for protein expression and surface display with Expi293F cells. Cell Biochem Funct 2023; 41:355-364. [PMID: 36864545 DOI: 10.1002/cbf.3787] [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: 11/23/2022] [Accepted: 02/18/2023] [Indexed: 03/04/2023]
Abstract
Compare with transient expression, stable cell lines generally have higher productivity and better quality for protein expression. However, selection of stable cell line is time-consuming and laborious. Here we describe an optimized selection method to achieve high-efficient stable cell pools with Expi293F suspension cells. This method only takes 2-3 weeks to generate stable cell pools with 2- to 10-fold higher productivity than transient gene expression (TGE). In fed-batch culture with Yeastolate, >1 g/L yield was achieved with our KTN0239-IgG stable cell pool in shaker flasks. This method can be also applied to efficiently display proteins on the cell surface.
Collapse
Affiliation(s)
- Chuan Chen
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - Zening Wang
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zehua Sun
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA.,Abound Bio, Pittsburgh, Pennsylvania, USA
| | - Wei Li
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - Dimiter S Dimitrov
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA.,Abound Bio, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Engineering Mechanical Strong Biomaterials Inspired by Structural Building Blocks in Nature. Chem Res Chin Univ 2023. [DOI: 10.1007/s40242-023-2357-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
32
|
Sui S, Wang H, Song J, Tai W. Development of a spermine lipid for transient antibody expression. Bioorg Med Chem 2023; 78:117114. [PMID: 36563514 DOI: 10.1016/j.bmc.2022.117114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Transient expression is the only way to quickly obtain a small scale of antibodies for biomedical research and therapeutic evaluation. The agents for transfecting the suspension cells, e.g. PEI or commercial agents, either lack efficiency or excessively expensive. Herein, a novel spermine-based lipid was developed and fabricated into a cationic liposome for antibody expression. This new transfection agent, designated as sperminoliposome, is feasible, cheap, and highly effective to produce antibodies. Compared to PEI, a 3 times higher yield of antibody was obtained by sperminoliposome during the transient expression of cetuximab in suspension 293F cells. Characterizations confirmed that the expressed antibody is fully functional and eligible for further research. Our study provides an effective tool for the rapid production of antibodies economically and feasibly.
Collapse
Affiliation(s)
- Shaowei Sui
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Hao Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Jiajie Song
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wanyi Tai
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China; Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
33
|
Milne JJ. Mixed-Mode Chromatography and Its Role in Monoclonal Antibody Purification. Methods Mol Biol 2023; 2699:15-29. [PMID: 37646991 DOI: 10.1007/978-1-0716-3362-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
As the biopharmaceutical industry matures and embraces process intensification methodologies allied to the emergence of newer personalized medicines, a key constant is the regulatory need to purify products that satisfy the criteria of safety, quality, and efficacy in each batch of released product destined for clinical use. Downstream processing operations and in particular chromatographic separations continue to play a key role in manufacturing strategies with the industry being well served by commercially available resins that provide different options to purify a particular target molecule of interest. In recent years, mixed-mode chromatography, a technique based on multimode interactions between ligands and proteins, had attracted much attention. This short review will discuss the concept and benefit of mixed-mode chromatography in purification strategies and specifically look at its application in the purification of IgG subtype monoclonal antibodies, a key product class in today's industry.
Collapse
Affiliation(s)
- John Joseph Milne
- National Institute for Bioprocessing Research and Training (NIBRT), Blackrock, Dublin, Ireland.
| |
Collapse
|
34
|
Oliviero C, Hinz SC, Grzeschik J, Hock B, Kolmar H, Hagens G. Cell Line Development Using Targeted Gene Integration into MAR-Rich Landing Pads for Stable Expression of Transgenes. Methods Mol Biol 2023; 2681:343-359. [PMID: 37405657 DOI: 10.1007/978-1-0716-3279-6_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Integration of a gene of interest (GOI) into the genome of mammalian cells is the first step of cell line development campaigns for the production of biotherapeutics. Besides random integration methods, targeted gene integration approaches have emerged as promising tools over the last few years. In addition to reducing heterogeneity within a pool of recombinant transfectants, this process can also facilitate shorter timelines of the current cell line development process. Herein, we describe protocols for generating host cell lines carrying matrix attachment region (MAR)-rich landing pads (LPs), including BxB1 recombination sites. These LP-containing cell lines allow for site-specific and simultaneous integration of multiple GOIs. The resulting transgene-expressing stable recombinant clones can be used for the production of mono- or multispecific antibodies.
Collapse
Affiliation(s)
- Claudia Oliviero
- Institute of Life Technologies, Haute Ecole d'Ingénierie HES-SO Valais Wallis, Sion, Switzerland
| | - Steffen C Hinz
- Institute of Life Technologies, Haute Ecole d'Ingénierie HES-SO Valais Wallis, Sion, Switzerland
| | | | - Björn Hock
- Aerium Therapeutics, Epalinges, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | - Gerrit Hagens
- Institute of Life Technologies, Haute Ecole d'Ingénierie HES-SO Valais Wallis, Sion, Switzerland.
| |
Collapse
|
35
|
Milne JJ. Scale-Up of Protein Purification: Downstream Processing Issues. Methods Mol Biol 2023; 2699:61-75. [PMID: 37646994 DOI: 10.1007/978-1-0716-3362-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Large-scale chromatography operations continue to occupy the central position in the overall strategy for downstream processing and purification of therapeutic protein products for human use. As the biopharmaceutical industry looks forward to embracing new therapeutic modalities such as viral vector-mediated gene therapy, it is becoming evident that chromatographic separations will be also be crucial for success in that discipline. The current industry focus on cell culture intensification strategies that can result in increased process efficiency and lower cost of goods is presenting challenges to the robustness and economics of chromatography processes. To ensure robust and reproducible commercial manufacturing strategies, there is always a mandate to increase the scale of chromatography unit operations that are typically developed and optimized in small-scale development trials. This chapter discusses the key factors in typical chromatography operations that need to be carefully considered and modeled during the process scale-up phase in order to maintain the purity, yield, and quality of a product purified at smaller scales.
Collapse
Affiliation(s)
- John Joseph Milne
- National Institute for Bioprocessing Research and Training (NIBRT), Blackrock, Dublin, Ireland.
| |
Collapse
|
36
|
Henehan GT, Ryan BJ, Kinsella GK. Approaches to Avoid Proteolysis During Protein Expression and Purification. Methods Mol Biol 2023; 2699:77-95. [PMID: 37646995 DOI: 10.1007/978-1-0716-3362-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
All cells contain proteases, which hydrolyze the peptide bonds between amino acids of a protein backbone. Typically, proteases are prevented from nonspecific proteolysis by regulation and by their physical separation into different subcellular compartments; however, this segregation is not retained during cell lysis, which is the initial step in any protein isolation procedure. Prevention of proteolysis during protein purification often takes the form of a two-pronged approach: first, inhibition of proteolysis in situ, followed by the early separation of the protease from the protein of interest via chromatographic purification. Protease inhibitors are routinely used to limit the effect of the proteases before they are physically separated from the protein of interest via column chromatography. In this chapter, commonly used approaches to reducing or avoiding proteolysis during protein expression and purification are reviewed.
Collapse
Affiliation(s)
- Gary T Henehan
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman, Dublin, Ireland
| | - Barry J Ryan
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman, Dublin, Ireland
| | - Gemma K Kinsella
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman, Dublin, Ireland.
| |
Collapse
|
37
|
Wysor SK, Hall KA, Marcus RK. Rapid metal speciation of cell culture media using reversed-phase separations and inductively coupled plasma optical emission spectrometry. Biotechnol Prog 2023; 39:e3311. [PMID: 36308722 DOI: 10.1002/btpr.3311] [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: 07/28/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022]
Abstract
Cell culture media metal content is critical in mammalian cell growth and monoclonal antibody productivity. The variability in metal concentrations has multiple sources of origin. As such, there is a need to analyze media before, during, and after production. Furthermore, it is not the simple presence of a given metal that can impact processes, but also their chemical form that is, speciation. To a first approximation, it is instructive to simply and quickly ascertain if the metals exist as inorganic (free metal) ions or are part of an organometallic complex (ligated). Here we present a simple workflow involving the capture of ligated metals on a fiber stationary phase with passage of the free ions to an inductively coupled plasma optical emission spectrometry for quantification; the captured species are subsequently eluted for quantification. This first level of speciation (free vs. ligated) can be informative towards sources of contaminant metal species and means to assess bioreactor processes.
Collapse
Affiliation(s)
- Sarah K Wysor
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, South Carolina, USA
| | - Katja A Hall
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, South Carolina, USA
| | - R Kenneth Marcus
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
38
|
Pandey Y, Dondapati SK, Wüstenhagen D, Kubick S. Cell-Free Synthesis and Electrophysiological Analysis of Multipass Voltage-Gated Ion Channels Tethered in Microsomal Membranes. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2023; 186:103-120. [PMID: 37640910 DOI: 10.1007/10_2023_228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Cell-free protein synthesis (CFPS) has emerged as a powerful tool for the rapid synthesis and analysis of various structurally and functionally distinct proteins. These include 'difficult-to-express' membrane proteins such as large multipass ion channel receptors. Owing to their membrane localization, eukaryotic CFPS supplemented with endoplasmic reticulum (ER)-derived microsomal vesicles has proven to be an efficient system for the synthesis of functional membrane proteins. Here we demonstrate the applicability of the eukaryotic cell-free systems based on lysates from the mammalian Chinese Hamster Ovary (CHO) and insect Spodoptera frugiperda (Sf21) cells. We demonstrate the efficiency of the systems in the de novo cell-free synthesis of the human cardiac ion channels: ether-a-go-go potassium channel (hERG) KV11.1 and the voltage-gated sodium channel hNaV1.5.
Collapse
Affiliation(s)
- Yogesh Pandey
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
- Institut für Biochemie und Biologie, University of Potsdam, Potsdam, OT Golm, Germany
| | - Srujan Kumar Dondapati
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany.
| | - Doreen Wüstenhagen
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
| | - Stefan Kubick
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
- Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
- Institute of Chemistry and Biochemistry-Biochemistry, Freie Universität Berlin, Berlin, Germany
- Faculty of Health Science, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, The Brandenburg Medical School Theodor Fontane and the University of Potsdam, Potsdam, Germany
| |
Collapse
|
39
|
Nandal J, Mihooliya KN, Verma H, Kalidas N, Ashish F, Mishra RPN, Sahoo DK. Evaluation of physicochemical and functional similarity of a new CHO derived anti-EGFR antibody P-mAb to its reference medicinal product. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:17-28. [DOI: 10.1080/21691401.2022.2028284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Affiliation(s)
- Jitender Nandal
- Biochemical Engineering Research and Process Development Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Kanti N. Mihooliya
- Biochemical Engineering Research and Process Development Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Himanshu Verma
- Biochemical Engineering Research and Process Development Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Nidhi Kalidas
- GNR Advanced Protein Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Fnu Ashish
- GNR Advanced Protein Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Ravi P. N. Mishra
- Biochemical Engineering Research and Process Development Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Debendra K. Sahoo
- Biochemical Engineering Research and Process Development Centre, CSIR-Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
40
|
Fuselli A, de Los Milagros Bürgi M, Kratje R, Prieto C. Generation and functional evaluation of novel monoclonal antibodies targeting glycosylated human stem cell factor. Appl Microbiol Biotechnol 2022; 106:8121-8137. [PMID: 36401641 DOI: 10.1007/s00253-022-12282-6] [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: 03/16/2022] [Revised: 10/31/2022] [Accepted: 11/05/2022] [Indexed: 11/21/2022]
Abstract
Human stem cell factor (hSCF) is an early-acting growth factor that promotes proliferation, differentiation, migration, and survival in several tissues. It plays a crucial role in hematopoiesis, gametogenesis, melanogenesis, intestinal motility, and in development and recovery of nervous and cardiovascular systems. Potential therapeutic applications comprise anemia treatment, mobilization of hematopoietic stem/progenitor cells to peripheral blood, and increasing gene transduction efficiency for gene therapy. Developing new tools to characterize recombinant hSCF in most native-like form as possible is crucial to understand the complexity of its in vivo functions and for improving its biotechnological applications. The soluble domain of hSCF was expressed in HEK293 cells. Highly purified rhSCF showed great molecular mass variability due to the presence of N- and O-linked carbohydrates, and it presented a 2.5-fold increase on proliferative activity compared to bacteria-derived hSCF. Three hybridoma clones producing monoclonal antibodies (mAbs) with high specificity for the glycoprotein were obtained. 1C4 and 2D3 mAbs were able to detect bacteria-derived and glycosylated rhSCF and demonstrated to be excellent candidates to develop a sandwich ELISA assay for rhSCF quantification, with detection limits of 0.18 and 0.07 ng/ml, respectively. Interestingly, 1A10 mAb only recognized glycosylated rhSCF, suggesting that sugar moieties might be involved in epitope recognition. 1A10 mAb showed the highest binding affinity, and it constituted the best candidate for immunodetection of the entire set rhSCF glycoforms in western blot assays, and for intracellular cytokine staining. Our work shows that combining glycosylated rhSCF expression with hybridoma technology is a powerful strategy to obtain specific suitable immunochemical assays and thus improve glycoprotein-producing bioprocesses. KEY POINTS: • Soluble glycosylated human SCF exerted improved proliferative activity on UT-7 cells. • Three mAbs with high specificity targeting glycosylated human SCF were obtained. • mAbs applications comprise sandwich ELISA, western blot, and immunofluorescence assays.
Collapse
Affiliation(s)
- Antonela Fuselli
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Cell Culture Laboratory, Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242 (S3000ZAA), Santa Fe, Argentina
| | - María de Los Milagros Bürgi
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Cell Culture Laboratory, Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242 (S3000ZAA), Santa Fe, Argentina
| | - Ricardo Kratje
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Cell Culture Laboratory, Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242 (S3000ZAA), Santa Fe, Argentina
| | - Claudio Prieto
- UNL, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Biotechnological Development Laboratory, Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242 (S3000ZAA), Santa Fe, Argentina.
- Cellargen Biotech S.R.L., Antonia Godoy 6369 (S3000ZAA), Santa Fe, Argentina.
| |
Collapse
|
41
|
The Effect of Sample Glucose Content on PNGase F-Mediated N-Glycan Release Analyzed by Capillary Electrophoresis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238192. [PMID: 36500281 PMCID: PMC9740771 DOI: 10.3390/molecules27238192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Protein therapeutics have recently gained high importance in general health care along with applied clinical research. Therefore, it is important to understand the structure-function relationship of these new generation drugs. Asparagine-bound carbohydrates represent an important critical quality attribute of therapeutic glycoproteins, reportedly impacting the efficacy, immunogenicity, clearance rate, stability, solubility, pharmacokinetics and mode of action of the product. In most instances, these linked N-glycans are analyzed in their unconjugated form after endoglycosidase-mediated release, e.g., PNGase F-mediated liberation. In this paper, first, N-glycan release kinetics were evaluated using our previously reported in-house produced 6His-PNGase F enzyme. The resulting deglycosylation products were quantified by sodium dodecyl sulfate capillary gel electrophoresis to determine the optimal digestion time. Next, the effect of sample glucose content was investigated as a potential endoglycosidase activity modifier. A comparative Michaelis-Menten kinetics study was performed between the 6His-PNGase F and a frequently employed commercial PNGase F product with and without the presence of glucose in the digestion reaction mixture. It was found that 1 mg/mL glucose in the sample activated the 6His-PNGase F enzyme, while did not affect the release efficiency of the commercial PNGase F. Capillary isoelectric focusing revealed subtle charge heterogeneity differences between the two endoglycosidases, manifested by the lack of extra acidic charge variants in the cIEF trace of the 6His-PNGase F enzyme, which might have possibly influenced the glucose-mediated enzyme activity differences.
Collapse
|
42
|
Rodríguez MC, Ceaglio N, Gugliotta A, Villarraza J, Garay E, Fuselli A, Gastaldi V, Tardivo MB, Antuña S, Fontana D, Prieto C. Design and optimization of an IgG human ELISA assay reactive to recombinant RBD SARS-CoV-2 protein. Appl Microbiol Biotechnol 2022; 106:7933-7948. [DOI: 10.1007/s00253-022-12254-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/17/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
|
43
|
Yang N, Zhang J, Xu M, Yi J, Wang Z, Wang Y, Chen C. Virus-like particles vaccines based on glycoprotein E0 and E2 of bovine viral diarrhea virus induce Humoral responses. Front Microbiol 2022; 13:1047001. [PMID: 36439839 PMCID: PMC9687372 DOI: 10.3389/fmicb.2022.1047001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 10/10/2022] [Indexed: 07/27/2023] Open
Abstract
Bovine viral diarrhea/mucosal disease (BVD/MD) is a viral infectious disease that seriously endangers the health of cattle herds and brings serious economic losses to the global cattle industry. Virus-like particles (VLPs) are empty shell structures without viral nucleic acid, which are similar to natural virus particles in morphology and structure. Because of their strong immunogenicity and biological activity, some of them have been used as vaccines in clinical trials. In this study, we developed a strategy to generate BVDV (E0 + E2, E2 + E2) VLPs using an insect baculovirus expression vector system (BEVS). The VLPs obtained were detected by immunofluorescence assay (IFA), western blotting analyses and transmission electron microscope (TEM), and the results showed that VLPs of high purity were obtained. Mice immunized with VLPs (15 μg) and Freund's adjuvant (100 μl) elicited higher BVDV-neutralizing antibody in comparison with Freund's adjuvant control (p < 0.0001), and even on day 21 or 35 post-prime immunization, the neutralizing antibody levels of mice immunized with E0 + E2 or E2 + E2 VLPs were significantly higher compared with inactivated vaccine (p < 0.05). A subsequent challenge reveals that the viral loads of livers, kidneys, spleens, lungs and small intestines were significantly lower compared with control (p < 0.0001), and the viral loads of mice immunized with E0 + E2 or E2 + E2 VLPs in the small intestines were significantly lower compared with inactivated vaccine (p < 0.05). Thus, VLPs are a promising candidate vaccine and warrants further clinical evaluation.
Collapse
Affiliation(s)
- Ningning Yang
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Jiangwei Zhang
- Intelligent Breeding of Livestock and Poultry, Tiemenguan Vocational and Technical College, Tiemenguan, China
| | - Mingguo Xu
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Jihai Yi
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Zhen Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yong Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Shihezi, China
- Key Laboratory of Control and Prevention of Animal Disease, Xinjiang Production & Construction Corps, Shihezi, China
- Co-Innovation Center for Zoonotic Infectious Diseases in the Western Region, Shihezi, China
| |
Collapse
|
44
|
Li Y, Champion JA. Self-assembling nanocarriers from engineered proteins: Design, functionalization, and application for drug delivery. Adv Drug Deliv Rev 2022; 189:114462. [PMID: 35934126 DOI: 10.1016/j.addr.2022.114462] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/09/2022] [Accepted: 07/15/2022] [Indexed: 01/24/2023]
Abstract
Self-assembling proteins are valuable building blocks for constructing drug nanocarriers due to their self-assembly behavior, monodispersity, biocompatibility, and biodegradability. Genetic and chemical modifications allow for modular design of protein nanocarriers with effective drug encapsulation, targetability, stimuli responsiveness, and in vivo half-life. Protein nanocarriers have been developed to deliver various therapeutic molecules including small molecules, proteins, and nucleic acids with proven in vitro and in vivo efficacy. This article reviews recent advances in protein nanocarriers that are not derived from natural protein nanostructures, such as protein cages or virus like particles. The protein nanocarriers described here are self-assembled from rationally or de novo designed recombinant proteins, as well as recombinant proteins complexed with other biomolecules, presenting properties that are unique from those of natural protein carriers. Design, functionalization, and therapeutic application of protein nanocarriers will be discussed.
Collapse
Affiliation(s)
- Yirui Li
- BioEngineering Program, Georgia Institute of Technology, United States
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, United States; BioEngineering Program, Georgia Institute of Technology, United States.
| |
Collapse
|
45
|
Rozov SM, Deineko EV. Increasing the Efficiency of the Accumulation of Recombinant Proteins in Plant Cells: The Role of Transport Signal Peptides. PLANTS (BASEL, SWITZERLAND) 2022; 11:2561. [PMID: 36235427 PMCID: PMC9572730 DOI: 10.3390/plants11192561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
The problem with increasing the yield of recombinant proteins is resolvable using different approaches, including the transport of a target protein to cell compartments with a low protease activity. In the cell, protein targeting involves short-signal peptide sequences recognized by intracellular protein transport systems. The main systems of the protein transport across membranes of the endoplasmic reticulum and endosymbiotic organelles are reviewed here, as are the major types and structure of the signal sequences targeting proteins to the endoplasmic reticulum and its derivatives, to plastids, and to mitochondria. The role of protein targeting to certain cell organelles depending on specific features of recombinant proteins and the effect of this targeting on the protein yield are discussed, in addition to the main directions of the search for signal sequences based on their primary structure. This knowledge makes it possible not only to predict a protein localization in the cell but also to reveal the most efficient sequences with potential biotechnological utility.
Collapse
|
46
|
Duan YH, Bian YL, Zhu JW. Generating Anti-TIGIT and CD155 Monoclonal Antibodies for Tumor Immunotherapy. PHARMACEUTICAL FRONTS 2022. [DOI: 10.1055/s-0042-1755454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
AbstractMany studies have confirmed that the human poliovirus receptor (PVR; CD155) is related to tumor cell migration, invasion, and thus tumor progression. A PVR receptor binds its ligand T cell Ig and the ITIM domain (TIGIT) to inhibit the function of T and NK cells, thereby allowing tumors to evade immune surveillance. In this study, two IgG1 monoclonal antibodies, anti-CD155 and anti-TIGIT, were expressed by the mammalian transient transfection system, then, antibody-dependent cell-mediated cytotoxicity, antibody-binding affinity, and antitumor efficacy were evaluated subsequently in vitro. In this work, protein A affinity chromatography was used for antibodies' purification. Analysis methods included Western blot, enzyme-linked immunosorbent assay, and flow cytometry. Our data suggested that both the two monoclonal antibodies have a purity of higher than 90%, and bound tightly to the antigen with dissociation constant (K
d) and 50% effective concentrations (EC50) below micromolar range. Most notably, these antibodies promote antitumor activity of immune cells in vitro. Therefore, our study laid down the foundation for subsequent in vivo experiments for further evaluation.
Collapse
Affiliation(s)
- Yu-Hang Duan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yan-lin Bian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jian-Wei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
47
|
Impact of SARS-CoV-2 RBD Mutations on the Production of a Recombinant RBD Fusion Protein in Mammalian Cells. Biomolecules 2022; 12:biom12091170. [PMID: 36139010 PMCID: PMC9496381 DOI: 10.3390/biom12091170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
SARS-CoV-2 receptor-binding domain (RBD) is a major target for the development of diagnostics, vaccines and therapeutics directed against COVID-19. Important efforts have been dedicated to the rapid and efficient production of recombinant RBD proteins for clinical and diagnostic applications. One of the main challenges is the ongoing emergence of SARS-CoV-2 variants that carry mutations within the RBD, resulting in the constant need to design and optimise the production of new recombinant protein variants. We describe here the impact of naturally occurring RBD mutations on the secretion of a recombinant Fc-tagged RBD protein expressed in HEK 293 cells. We show that mutation E484K of the B.1.351 variant interferes with the proper disulphide bond formation and folding of the recombinant protein, resulting in its retention into the endoplasmic reticulum (ER) and reduced protein secretion. Accumulation of the recombinant B.1.351 RBD-Fc fusion protein in the ER correlated with the upregulation of endogenous ER chaperones, suggestive of the unfolded protein response (UPR). Overexpression of the chaperone and protein disulphide isomerase PDIA2 further impaired protein secretion by altering disulphide bond formation and increasing ER retention. This work contributes to a better understanding of the challenges faced in producing mutant RBD proteins and can assist in the design of optimisation protocols.
Collapse
|
48
|
Nagata S, Yamasaki M, Kuroishi N, Kitamura K. Development of Long-Acting Human Adrenomedullin Fc-Fusion Proteins. BIOLOGY 2022; 11:biology11071074. [PMID: 36101452 PMCID: PMC9312564 DOI: 10.3390/biology11071074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 12/01/2022]
Abstract
Simple Summary Adrenomedullin (AM) is a hypotensive peptide hormone that exerts anti-inflammatory effects and is involved in wound healing and embryogenesis. However, treatment requires continuous administration as the half-life of native AM is short in blood. To resolve this, we developed four human IgG1 and IgG4 Fc-fusion proteins containing full-length hAM or hAM residues 6-52 using mammalian cells. The Fc-AM produced were amidated and in the active form. All Fc-AMs stimulated cAMP production in HEK-293 cells stably expressing the AM1 receptor. The activities of IgG1-AM (6-52) and IgG4-AM (6-52) were higher than those of IgG1-AM and IgG4-AM. Sufficient concentrations of IgG1-AM (6-52) and IgG4-AM (6-52) were observed in blood 14 days after a single subcutaneous administration. Furthermore, after IgG1-AM (6-52) or IgG4-AM (6-52) administration, tissue transfer to the kidney and small intestine was observed. Treatment with IgG4-AM (6-52) inhibited blood pressure increase in spontaneously hypertensive rats. Fc-AM produced from mammalian cells can be easily prepared and might be an effective novel therapeutic agent. Abstract (1) Background: Human adrenomedullin (hAM) is a hypotensive peptide hormone that exerts powerful anti-inflammatory effects. AM also had therapeutic effects in various animal experimental models of disease. However, treatment required continuous administration as the half-life of native AM is short in blood. To resolve this, we developed four human IgG1 and IgG4 Fc-fusion proteins containing full-length hAM or hAM residues 6-52. (2) Methods: We used mammalian cells to produce recombinant Fc-AM derivatives and tested the pharmacokinetics and biological activity of Fc-AM. (3) Results: We developed four Fc-fusion AMs (Fc-AM), which are long-acting AM derivatives in mammalian cells. Fc-AM had a prolonged half-life in blood and retained its ability to bind to the AM1 receptor. Fc-AM (6-52) induced higher cAMP levels for the receptor than Fc-AM. After the administration of IgG1-AM (6-52) or IgG4-AM (6-52) to rats, tissue transfer to the kidney and small intestine was observed. In addition, treatment with IgG4-AM (6-52) inhibited blood pressure increase in spontaneously hypertensive rats. (4) Conclusions: Fc-AM produced from mammalian cells can be easily prepared and might be an effective novel therapeutic agent.
Collapse
|
49
|
Srila W, Baumann M, Borth N, Yamabhai M. Codon and signal peptide optimization for therapeutic antibody production from Chinese hamster ovary (CHO) cell. Biochem Biophys Res Commun 2022; 622:157-162. [PMID: 35863090 DOI: 10.1016/j.bbrc.2022.06.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/22/2022] [Indexed: 11/02/2022]
Affiliation(s)
- Witsanu Srila
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Martina Baumann
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, Austria
| | - Nicole Borth
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria.
| | - Montarop Yamabhai
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, Thailand.
| |
Collapse
|
50
|
Li Q, Yan RF, Yang YX, Mi CL, Jia YL, Wang TY. Stabilizing and Anti-Repressor Elements Effectively Increases Transgene Expression in Transfected CHO Cells. Front Bioeng Biotechnol 2022; 10:840600. [PMID: 35721852 PMCID: PMC9199445 DOI: 10.3389/fbioe.2022.840600] [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: 12/22/2021] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Chinese hamster ovary (CHO) cells are currently the most widely used host cells for recombinant therapeutic protein (RTP) production. Currently, the RTP yields need to increase further to meet the market needs and reduce costs. In this study, three stabilizing and anti-repressor (SAR) elements from the human genome were selected, including human SAR7, SAR40, and SAR44 elements. SAR elements were cloned upstream of the promoter in the eukaryotic vector, followed by transfection into CHO cells, and were screened under G418 pressure. Flow cytometry was used to detect enhanced green fluorescent protein (eGFP) expression levels. The gene copy numbers and mRNA expression levels were determined through quantitative real-time PCR. Furthermore, the effect of the stronger SAR elements on adalimumab was investigated. The results showed that transgene expression levels in the SAR-containing vectors were higher than that of the control vector, and SAR7 and SAR40 significantly increased and maintained the long-term expression of the transgene in CHO cells. In addition, the transgene expression level increase was related with gene copy numbers and mRNA expression levels. Collectively, SAR elements can enhance the transgene expression and maintain the long-term expression of a transgene in transfected CHO cells, which may be used to increase recombinant protein production in CHO cells.
Collapse
Affiliation(s)
- Qin Li
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China.,International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| | - Rui-Fang Yan
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Yong-Xiao Yang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| | - Chun-Liu Mi
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China.,International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yan-Long Jia
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| | - Tian-Yun Wang
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China.,International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|