1
|
Tsukidate T, Sahoo A, Pendyala G, Yang RS, Welch J, Madabhushi S, Li X. Discovery of Chemical Tools for Polysorbate-Degradative Enzyme Control in the Biopharmaceutical Upstream Process via Multi-Omic Profiling of Host Cell Clones. ACS Chem Biol 2025. [PMID: 40249937 DOI: 10.1021/acschembio.5c00081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2025]
Abstract
Host cell proteins are process-related impurities in biotherapeutics and can potentially pose risks to patient safety and product quality. Specifically, certain host cell-derived enzymes, including lipases, can degrade the formulation excipient polysorbate (PS) in biopharmaceutical formulations, affecting drug product stability in liquid formulations. We leveraged multiomics approaches, including transcriptomics, proteomics, and activity-based protein profiling (ABPP), to identify mechanisms that regulate PS-degradative enzyme (PSDE) abundance and to develop strategies for their control. Comparative multiomics analysis of two monoclonal antibody (mAb)-producing host cell clones revealed differential lipase profiles at the mRNA, protein, and enzyme activity levels and associated increased lipase activity with upregulated lipid catabolic pathways such as the fatty acid beta oxidation pathway. Further, for the first time in the literature, we identified peroxisome proliferator-activated receptor γ (PPARγ) as a key regulator of PSDEs in manufacturing Chinese Hamster Ovary (CHO) cells. Downregulation of the PPARγ pathway with its antagonists resulted in a selective reduction of PSDE levels and improved PS stability without compromising mAb productivity or quality. This study highlights the potential of PPARγ modulators as chemical tools for PSDE control at the gene regulation level, offering significant implications for biopharmaceutical process development and control.
Collapse
Affiliation(s)
- Taku Tsukidate
- Analytical Research & Development Mass Spectrometry, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Ansuman Sahoo
- Biologics Process Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Geetanjali Pendyala
- Biologics Process Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Rong-Sheng Yang
- Analytical Research & Development Mass Spectrometry, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Jonathan Welch
- Biologics Analytical Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Sri Madabhushi
- Biologics Process Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Xuanwen Li
- Analytical Research & Development Mass Spectrometry, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, New Jersey 07065, United States
| |
Collapse
|
2
|
Sahoo A, Tsukiadate T, Lin B, Kotzbauer E, Houser J, Patel M, Li X, Madabhushi SR. Proteomics Reveals Distinctive Host Cell Protein Expression Patterns in Fed-Batch and Perfusion Cell Culture Processes. Biotechnol J 2025; 20:e202400567. [PMID: 39834099 PMCID: PMC11747259 DOI: 10.1002/biot.202400567] [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: 09/17/2024] [Revised: 11/20/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
Chinese hamster ovary (CHO) cells are widely used to produce recombinant proteins, including monoclonal antibodies (mAbs), through various process modes. While fed-batch (FB) processes have been the standard, a shift toward high-density perfusion processes is being driven by increased productivity, flexible facility footprints, and lower costs. Ensuring the clearance of process-related impurities, such as host cell proteins (HCPs), is crucial in biologics manufacturing. Although purification processes remove most impurities, integrated strategies are being developed to enhance clearance of some high-risk HCPs. Current understanding of HCP expression dynamics in cell culture is limited. This study utilized data-independent acquisition (DIA) proteomics to compare the proteomic profiles of cell culture supernatants from 14 FB clones and three perfusion clones, all expressing the same mAb from the same host cell line. Results showed that perfusion processes enhance cell growth and productivity, exhibiting distinct proteomic profiles compared to FB processes. Perfusion processes also maintain a more comparable HCP abundance profile across clones, especially for 46 problematic HCPs monitored. Cluster analysis of FB proteomics revealed distinct abundance patterns and correlations with process parameters. Differential abundance analysis identified significant protein differences between the two processes. This is the first extensive study characterizing HCPs expressed by clones under different process modes. Further research could lead to strategies for preventing or managing problematic HCPs in biologics manufacturing.
Collapse
Affiliation(s)
- Ansuman Sahoo
- Biologics Process Research & DevelopmentMerck & Co., Inc.RahwayNew JerseyUSA
| | - Taku Tsukiadate
- Analytical Research & Development Mass SpectrometryMerck & Co., Inc.RahwayNew JerseyUSA
| | - Bor‐Ruei Lin
- Biologics Process Research & DevelopmentMerck & Co., Inc.RahwayNew JerseyUSA
| | - Erin Kotzbauer
- Biologics Process Research & DevelopmentMerck & Co., Inc.RahwayNew JerseyUSA
| | - Jason Houser
- Biologics Process Research & DevelopmentMerck & Co., Inc.RahwayNew JerseyUSA
| | - Misaal Patel
- Biologics Process Research & DevelopmentMerck & Co., Inc.RahwayNew JerseyUSA
| | - Xuanwen Li
- Analytical Research & Development Mass SpectrometryMerck & Co., Inc.RahwayNew JerseyUSA
| | | |
Collapse
|
3
|
Sahoo A, Devenney K, Nicolas MA, Zill N, Gupta B, Madabhushi SR. Beyond viability: Advancing CHO cell culture process strategies to modulate host cell protein levels. N Biotechnol 2024; 84:128-139. [PMID: 39481535 DOI: 10.1016/j.nbt.2024.10.003] [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: 06/21/2024] [Revised: 09/25/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Chinese Hamster Ovary (CHO) cells are widely utilized in bioprocessing industry for monoclonal antibody (mAb) production. In many instances, challenges persist in achieving sufficient clearance of Host Cell Proteins (HCPs) in the final drug substance. While purification strategies usually offer substantial HCP clearance, certain "problematic" HCPs, particularly lipases, continue to pose significant challenges. This study investigates the accumulation of various "problematic" HCPs in CHO cell culture using transcriptomics, revealing correations between cell culture parameters and HCP level. Contrary to conventional assumptions, viability alone does not reliably predict HCP levels, with factors such as clone selection, host cell line choice, media and feed compositions significantly influencing HCP accumulation. Leveraging transcriptomics-based approaches, we demonstrate the potential of upstream process control strategies to mitigate HCP presence and improve biologic product quality. Our findings underscore the importance of considering diverse cell culture parameters in bioprocess optimization to ensure product stability and quality. While promising, further research is needed to elucidate the mechanisms underlying HCP release and propagation through downstream processing stages, emphasizing the necessity of a comprehensive integrated approach to HCP control in biologics production.
Collapse
Affiliation(s)
- Ansuman Sahoo
- Biologics Process Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, NJ 07065, United States
| | - Kyle Devenney
- Biologics Process Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, NJ 07065, United States
| | - Marcus-Allen Nicolas
- Biologics Process Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, NJ 07065, United States
| | - Nicholas Zill
- Biologics Process Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, NJ 07065, United States
| | - Balrina Gupta
- Biologics Process Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, NJ 07065, United States
| | - Sri Ranganayaki Madabhushi
- Biologics Process Research & Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, NJ 07065, United States.
| |
Collapse
|
4
|
Ito T, Lutz H, Tan L, Wang B, Tan J, Patel M, Chen L, Tsunakawa Y, Park B, Banerjee S. Host cell proteins in monoclonal antibody processing: Control, detection, and removal. Biotechnol Prog 2024; 40:e3448. [PMID: 38477405 DOI: 10.1002/btpr.3448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024]
Abstract
Host cell proteins (HCPs) are process-related impurities in a therapeutic protein expressed using cell culture technology. This review presents biopharmaceutical industry trends in terms of both HCPs in the bioprocessing of monoclonal antibodies (mAbs) and the capabilities for HCP clearance by downstream unit operations. A comprehensive assessment of currently implemented and emerging technologies in the manufacturing processes with extensive references was performed. Meta-analyses of published downstream data were conducted to identify trends. Improved analytical methods and understanding of "high-risk" HCPs lead to more robust manufacturing processes and higher-quality therapeutics. The trend of higher cell density cultures leads to both higher mAb expression and higher HCP levels. However, HCP levels can be significantly reduced with improvements in operations, resulting in similar concentrations of approx. 10 ppm HCPs. There are no differences in the performance of HCP clearance between recent enhanced downstream operations and traditional batch processing. This review includes best practices for developing improved processes.
Collapse
Affiliation(s)
- Takao Ito
- Life Science, Process Solutions, Merck Ltd. (An Affiliate of Merck KGaA, Darmstadt, Germany), Tokyo, Japan
| | - Herb Lutz
- Independent Consultant, Sudbury, Massachusetts, USA
| | - Lihan Tan
- Life Science Services, Sigma-Aldrich Pte Ltd, Singapore, Singapore
| | - Bin Wang
- Life Science, Process Solutions, Merck Chemicals (Shanghai) Co. Ltd. (An Affiliate of Merck KGaA Darmstadt, Germany), Shanghai, China
| | - Janice Tan
- Life Science, Process Solutions, Merck Pte Ltd. (An Affiliate of Merck KGaA, Darmstadt, Germany), Singapore
| | - Masum Patel
- Life Science, Process Solutions, Merck Life Sciences Pvt. Ltd. (An Affiliate of Merck KGaA, Darmstadt, Germany), Bangalore, India
| | - Lance Chen
- Life Science, Process Solutions, Merck Pte Ltd. (An Affiliate of Merck KGaA, Darmstadt, Germany), Singapore
| | - Yuki Tsunakawa
- Life Science, Process Solutions, Merck Ltd. (An Affiliate of Merck KGaA, Darmstadt, Germany), Tokyo, Japan
| | - Byunghyun Park
- Life Science, Process Solutions, Merck Ltd. (An Affiliate of Merck KGaA, Darmstadt, Germany), Seoul, South Korea
| | - Subhasis Banerjee
- Life Science, Process Solutions, Merck Life Sciences Pvt. Ltd. (An Affiliate of Merck KGaA, Darmstadt, Germany), Bangalore, India
| |
Collapse
|
5
|
Achleitner L, Winter M, Aguilar PP, Lingg N, Jungbauer A, Klausberger M, Satzer P. Robust and resource-efficient production process suitable for large-scale production of baculovirus through high cell density seed train and optimized infection strategy. N Biotechnol 2024; 80:46-55. [PMID: 38302001 DOI: 10.1016/j.nbt.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
The aim of this study was the development of a scalable production process for high titer (108 pfu/mL and above) recombinant baculovirus stocks with low cell line-derived impurities for the production of virus-like particles (VLP). To achieve this, we developed a high cell density (HCD) culture for low footprint cell proliferation, compared different infection strategies at multiplicity of infection (MOI) 0.05 and 0.005, different infection strategies and validated generally applicable harvest criteria of cell viability ≤ 80%. We also investigated online measurable parameters to observe the baculovirus production. The infection strategy employing a very low virus inoculum of MOI 0.005 and a 1:2 dilution with fresh medium one day after infection proved to be the most resource efficient. There, we achieved higher cell-specific titers and lower host cell protein concentrations at harvest than other tested infection strategies with the same MOI, while saving half of the virus stock for infecting the culture compared to other tested infection strategies. HCD culture by daily medium exchange was confirmed as suitable for seed train propagation, infection, and baculovirus production, equally efficient as the conventionally propagated seed train. Online measurable parameters for cell concentration and average cell diameter were found to be effective in monitoring the production process. The study concluded that a more efficient VLP production process in large scale can be achieved using this virus stock production strategy, which could also be extended to produce other proteins or extracellular vesicles with the baculovirus expression system.
Collapse
Affiliation(s)
- Lena Achleitner
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Martina Winter
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Patricia Pereira Aguilar
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Nico Lingg
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Alois Jungbauer
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Miriam Klausberger
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Peter Satzer
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
6
|
Drobnjakovic M, Hart R, Kulvatunyou BS, Ivezic N, Srinivasan V. Current challenges and recent advances on the path towards continuous biomanufacturing. Biotechnol Prog 2023; 39:e3378. [PMID: 37493037 DOI: 10.1002/btpr.3378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/13/2023] [Accepted: 06/21/2023] [Indexed: 07/27/2023]
Abstract
Continuous biopharmaceutical manufacturing is currently a field of intense research due to its potential to make the entire production process more optimal for the modern, ever-evolving biopharmaceutical market. Compared to traditional batch manufacturing, continuous bioprocessing is more efficient, adjustable, and sustainable and has reduced capital costs. However, despite its clear advantages, continuous bioprocessing is yet to be widely adopted in commercial manufacturing. This article provides an overview of the technological roadblocks for extensive adoptions and points out the recent advances that could help overcome them. In total, three key areas for improvement are identified: Quality by Design (QbD) implementation, integration of upstream and downstream technologies, and data and knowledge management. First, the challenges to QbD implementation are explored. Specifically, process control, process analytical technology (PAT), critical process parameter (CPP) identification, and mathematical models for bioprocess control and design are recognized as crucial for successful QbD realizations. Next, the difficulties of end-to-end process integration are examined, with a particular emphasis on downstream processing. Finally, the problem of data and knowledge management and its potential solutions are outlined where ontologies and data standards are pointed out as key drivers of progress.
Collapse
Affiliation(s)
- Milos Drobnjakovic
- Systems Integration Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Roger Hart
- National Institute for Innovation in Manufacturing Biopharmaceuticals, Newark, New Jersey, USA
| | - Boonserm Serm Kulvatunyou
- Systems Integration Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Nenad Ivezic
- Systems Integration Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Vijay Srinivasan
- Systems Integration Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| |
Collapse
|
7
|
Hamaker NK, Min L, Lee KH. Comprehensive Assessment of Host Cell Protein Expression after Extended Culture and Bioreactor Production of CHO Cell Lines. Biotechnol Bioeng 2022; 119:2221-2238. [PMID: 35508759 DOI: 10.1002/bit.28128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 11/12/2022]
Abstract
The biomanufacturing industry is advancing toward continuous processes that will involve longer culture durations and older cell ages. These upstream trends may bring unforeseen challenges for downstream purification due to fluctuations in host cell protein (HCP) levels. To understand the extent of HCP expression instability exhibited by Chinese hamster ovary (CHO) cells over these time scales, an industry-wide consortium collaborated to develop a study to characterize age-dependent changes in HCP levels across 30, 60, and 90 cell doublings, representing a period of approximately 60 days. A monoclonal antibody (mAb)-producing cell line with bulk productivity up to 3 g/L in a bioreactor was aged in parallel with its parental CHO-K1 host. Subsequently, both cell types at each age were cultivated in an automated bioreactor system to generate harvested cell culture fluid (HCCF) for HCP analysis. More than 1,500 HCPs were quantified using complementary proteomic techniques, two-dimensional electrophoresis (2DE) and liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). While up to 13% of proteins showed variable expression with age, more changes were observed when comparing between the two cell lines with up to 47% of HCPs differentially expressed. A small subset (50 HCPs) with age-dependent expression were previously reported to be problematic as high-risk and/or difficult-to-remove impurities; however, the vast majority of these were down-regulated with age. Our findings suggest that HCP expression changes over this time scale may not be as dramatic and pose as great of a challenge to downstream processing as originally expected but that monitoring of variably expressed problematic HCPs remains critical. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nathaniel K Hamaker
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware
| | - Lie Min
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware
| |
Collapse
|
8
|
Park SY, Egan S, Cura AJ, Aron KL, Xu X, Zheng M, Borys M, Ghose S, Li Z, Lee K. Untargeted proteomics reveals upregulation of stress response pathways during CHO-based monoclonal antibody manufacturing process leading to disulfide bond reduction. MAbs 2021; 13:1963094. [PMID: 34424810 PMCID: PMC8386704 DOI: 10.1080/19420862.2021.1963094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Monoclonal antibody (mAb) interchain disulfide bond reduction can cause a loss of function and negatively impact the therapeutic’s efficacy and safety. Disulfide bond reduction has been observed at various stages during the manufacturing process, including processing of the harvested material. The factors and mechanisms driving this phenomenon are not fully understood. In this study, we examined the host cell proteome as a potential factor affecting the susceptibility of a mAb to disulfide bond reduction in the harvested cell culture fluid (HCCF). We used untargeted liquid-chromatography-mass spectrometry-based proteomics experiments in conjunction with a semi-automated protein identification workflow to systematically compare Chinese hamster ovary (CHO) cell protein abundances between bioreactor conditions that result in reduction-susceptible and reduction-free HCCF. Although the growth profiles and antibody titers of these two bioreactor conditions were indistinguishable, we observed broad differences in host cell protein (HCP) expression. We found significant differences in the abundance of glycolytic enzymes, key protein reductases, and antioxidant defense enzymes. Multivariate analysis of the proteomics data determined that upregulation of stress-inducible endoplasmic reticulum (ER) and other chaperone proteins is a discriminatory characteristic of reduction-susceptible HCP profiles. Overall, these results suggest that stress response pathways activated during bioreactor culture increase the reduction-susceptibility of HCCF. Consequently, these pathways could be valuable targets for optimizing culture conditions to improve protein quality.
Collapse
Affiliation(s)
- Seo-Young Park
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, USA.,School of Chemical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Susan Egan
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Anthony J Cura
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Kathryn L Aron
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Xuankuo Xu
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Mengyuan Zheng
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Michael Borys
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Sanchayita Ghose
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Zhengjian Li
- Biologics Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, USA
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, USA
| |
Collapse
|
9
|
Khanal O, Lenhoff AM. Developments and opportunities in continuous biopharmaceutical manufacturing. MAbs 2021; 13:1903664. [PMID: 33843449 PMCID: PMC8043180 DOI: 10.1080/19420862.2021.1903664] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/25/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Today's biologics manufacturing practices incur high costs to the drug makers, which can contribute to high prices for patients. Timely investment in the development and implementation of continuous biomanufacturing can increase the production of consistent-quality drugs at a lower cost and a faster pace, to meet growing demand. Efficient use of equipment, manufacturing footprint, and labor also offer the potential to improve drug accessibility. Although technological efforts enabling continuous biomanufacturing have commenced, challenges remain in the integration, monitoring, and control of traditionally segmented unit operations. Here, we discuss recent developments supporting the implementation of continuous biomanufacturing, along with their benefits.
Collapse
Affiliation(s)
- Ohnmar Khanal
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Abraham M. Lenhoff
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| |
Collapse
|
10
|
Abstract
In recent years process modelling has become an established method which generates digital twins of manufacturing plant operation with the aid of numerically solved process models. This article discusses the benefits of establishing process modelling, in-house or by cooperation, in order to support the workflow from process development, piloting and engineering up to manufacturing. The examples are chosen from the variety of botanicals and biologics manufacturing thus proving the broad applicability from variable feedstock of natural plant extracts of secondary metabolites to fermentation of complex molecules like mAbs, fragments, proteins and peptides.Consistent models and methods to simulate whole processes are available. To determine the physical properties used as model parameters, efficient laboratory-scale experiments are implemented. These parameters are case specific since there is no database for complex molecules of biologics and botanicals in pharmaceutical industry, yet.Moreover, Quality-by-Design approaches, demanded by regulatory authorities, are integrated within those predictive modelling procedures. The models could be proven to be valid and predictive under regulatory aspects. Process modelling does earn its money from the first day of application. Process modelling is a key-enabling tool towards cost-efficient digitalization in chemical-pharmaceutical industries.
Collapse
|
11
|
Kruse T, Kampmann M, Greller G. Aqueous Two‐Phase Extraction of Monoclonal Antibodies from High Cell Density Cell Culture. CHEM-ING-TECH 2020. [DOI: 10.1002/cite.202000017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Thomas Kruse
- Sartorius Stedim Biotech GmbH, BioProcessing August-Spindler-Straße 11 37079 Göttingen Germany
| | - Markus Kampmann
- Sartorius Stedim Biotech GmbH, BioProcessing August-Spindler-Straße 11 37079 Göttingen Germany
| | - Gerhard Greller
- Sartorius Stedim Biotech GmbH, BioProcessing August-Spindler-Straße 11 37079 Göttingen Germany
| |
Collapse
|
12
|
Kruse T, Kampmann M, Rüddel I, Greller G. An alternative downstream process based on aqueous two-phase extraction for the purification of monoclonal antibodies. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
Xu J, Xu X, Huang C, Angelo J, Oliveira CL, Xu M, Xu X, Temel D, Ding J, Ghose S, Borys MC, Li ZJ. Biomanufacturing evolution from conventional to intensified processes for productivity improvement: a case study. MAbs 2020; 12:1770669. [PMID: 32425110 PMCID: PMC7531520 DOI: 10.1080/19420862.2020.1770669] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Process intensification has shown great potential to increase productivity and reduce costs in biomanufacturing. This case study describes the evolution of a manufacturing process from a conventional processing scheme at 1000-L scale (Process A, n = 5) to intensified processing schemes at both 1000-L (Process B, n = 8) and 2000-L scales (Process C, n = 3) for the production of a monoclonal antibody by a Chinese hamster ovary cell line. For the upstream part of the process, we implemented an intensified seed culture scheme to enhance cell densities at the seed culture step (N-1) prior to the production bioreactor (N) by using either enriched N-1 seed culture medium for Process B or by operating the N-1 step in perfusion mode for Process C. The increased final cell densities at the N-1 step allowed for much higher inoculation densities in the production bioreactor operated in fed-batch mode and substantially increased titers by 4-fold from Process A to B and 8-fold from Process A to C, while maintaining comparable final product quality. Multiple changes were made to intensify the downstream process to accommodate the increased titers. New high-capacity resins were implemented for the Protein A and anion exchange chromatography (AEX) steps, and the cation exchange chromatography (CEX) step was changed from bind-elute to flow-through mode for the streamlined Process B. Multi-column chromatography was developed for Protein A capture, and an integrated AEX-CEX pool-less polishing steps allowed semi-continuous Process C with increased productivity as well as reductions in resin requirements, buffer consumption, and processing times. A cost-of-goods analysis on consumables showed 6.7–10.1 fold cost reduction from the conventional Process A to the intensified Process C. The hybrid-intensified process described here is easy to implement in manufacturing and lays a good foundation to develop a fully continuous manufacturing with even higher productivity in the future.
Collapse
Affiliation(s)
- Jianlin Xu
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | - Xuankuo Xu
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | - Chao Huang
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | - James Angelo
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | | | - Mengmeng Xu
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | - Xia Xu
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | - Deniz Temel
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | - Julia Ding
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | - Sanchayita Ghose
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | - Michael C Borys
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| | - Zheng Jian Li
- Global Product Development and Supply, Bristol-Myers Squibb Company , Devens, MA, USA
| |
Collapse
|
14
|
Singh SK, Mishra A, Yadav D, Budholiya N, Rathore AS. Understanding the mechanism of copurification of “difficult to remove” host cell proteins in rituximab biosimilar products. Biotechnol Prog 2020; 36:e2936. [DOI: 10.1002/btpr.2936] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/01/2019] [Accepted: 10/22/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Sumit K. Singh
- Department of Chemical EngineeringIndian Institute of Technology Hauz Khas New Delhi India
| | - Avinash Mishra
- Department of Chemical EngineeringIndian Institute of Technology Hauz Khas New Delhi India
| | - Divyanshi Yadav
- Department of Chemical EngineeringIndian Institute of Technology Hauz Khas New Delhi India
| | - Niharika Budholiya
- Department of Chemical EngineeringIndian Institute of Technology Hauz Khas New Delhi India
| | - Anurag S. Rathore
- Department of Chemical EngineeringIndian Institute of Technology Hauz Khas New Delhi India
| |
Collapse
|
15
|
Metzger KFJ, Voloshin A, Schillinger H, Kühnel H, Maurer M. Adsorptive filtration: A case study for early impurity reduction in an Escherichia coli production process. Biotechnol Prog 2019; 36:e2948. [PMID: 31837191 DOI: 10.1002/btpr.2948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/22/2019] [Accepted: 12/08/2019] [Indexed: 11/10/2022]
Abstract
Primary recovery of intracellular products from Escherichia coli requires cell disruption which leads to a massive release of process-related impurities burdening subsequent downstream process (DSP) unit operations. Especially, DNA and endotoxins challenge purification operations due to their size and concentrations. Consequently, an early reduction in impurities will not only simplify the production process but also increase robustness while alleviating the workload afterward. In the present work, we studied the proof of concept whether a nonwoven anion exchange filter material decreases soluble impurities immediately at the clarification step of E. coli DSP. In a first attempt, endotoxin burden was reduced by 4.6-fold and the DNA concentration by 3.6-fold compared to conventional depth filtration. A design of experiment for the adsorptive filtration approach was carried out to analyze the influence of different critical process parameters (CPPs) on impurity reduction. We showed that depending on the CPPs chosen, a DNA lowering of more than 3 log values, an endotoxin decrease of approximately 7 logs, and a minor HCP clearance of at least 0.3 logs could be achieved. Thus, we further revealed a chromatography column protecting effect when using adsorptive filtration beforehand.
Collapse
Affiliation(s)
- Karl F J Metzger
- Life Sciences, University of Applied Sciences Campus Vienna, Wien, AT, Austria.,Bioprocess Engineering, Austrian Centre of Industrial Biotechnology, Wien, AT, Austria.,Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Wien, AT, Austria
| | - Alexei Voloshin
- 3M Company, Separation and Purification Sciences Division, 3M Center, Saint Paul, Minnesota
| | - Harald Schillinger
- Life Sciences, University of Applied Sciences Campus Vienna, Wien, AT, Austria.,3M Österreich, 3M Separation and Purification Sciences Division, Wien, AT, Austria
| | - Harald Kühnel
- Life Sciences, University of Applied Sciences Campus Vienna, Wien, AT, Austria
| | - Michael Maurer
- Life Sciences, University of Applied Sciences Campus Vienna, Wien, AT, Austria.,Bioprocess Engineering, Austrian Centre of Industrial Biotechnology, Wien, AT, Austria
| |
Collapse
|
16
|
Kruse T, Schmidt A, Kampmann M, Strube J. Integrated Clarification and Purification of Monoclonal Antibodies by Membrane Based Separation of Aqueous Two-Phase Systems. Antibodies (Basel) 2019; 8:antib8030040. [PMID: 31544846 PMCID: PMC6784141 DOI: 10.3390/antib8030040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 11/16/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAb) are used for the treatment of numerous serious diseases, which have led to an increasing demand over the last decades. Increased cell density and mAb titer of the cultivation broth lead to great challenges for the subsequent clarification and capture operations in the downstream process. As an alternative approach to the conventional downstream process, a selective mAb extraction via an aqueous two-phase system (ATPS) directly from the cultivation broth of a mAb producing industrial relevant chinese hamster ovary (CHO) cell line was investigated. An efficient purification of the mAb was accomplished by the ATPS composition. The phase separation was realized by a newly developed membrane based phase separator. Moreover, a complete cell removal was integrated into this process by the used membrane. A selectivity between both phases was achieved by membrane modification. Yields up to 93% in the light phase and removal of process related impurities were obtained after aqueous two-phase extraction (ATPE). Phase separation performance as well as contact angles on the membrane were characterized for different ATPS. ATPE directly from the cultivation broth in combination with the new membrane based phase separation led to a mAb yield of 78% with a simultaneous reduction of deoxyribonucleic acid (DNA) and host cell protein (HCP) load.
Collapse
Affiliation(s)
- Thomas Kruse
- Institute for Separation and Process Technology, Clausthal University of Technology, Leibnizstraße 15, 38678 Clausthal-Zellerfeld, Germany
- Sartorius Stedim Biotech GmbH, August Spindler Straße 11, 37079 Göttingen, Germany
| | - Axel Schmidt
- Institute for Separation and Process Technology, Clausthal University of Technology, Leibnizstraße 15, 38678 Clausthal-Zellerfeld, Germany
| | - Markus Kampmann
- Sartorius Stedim Biotech GmbH, August Spindler Straße 11, 37079 Göttingen, Germany
| | - Jochen Strube
- Institute for Separation and Process Technology, Clausthal University of Technology, Leibnizstraße 15, 38678 Clausthal-Zellerfeld, Germany.
| |
Collapse
|
17
|
Distinct and Quantitative Validation Method for Predictive Process Modeling with Examples of Liquid-Liquid Extraction Processes of Complex Feed Mixtures. Processes (Basel) 2019. [DOI: 10.3390/pr7050298] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
As of today, industrial process development for liquid-liquid extraction and scale-up of extraction columns is based on an experimental procedure that requires tests in pilot-scale. This methodology consumes large amounts of material and time and the utilized scale-up equations are crude estimates including considerable safety margins. This approach is practical for well-known systems or low-value products coupled with high production scale, where such a scale-up methodology has less impact on the overall profitability. However, for new high-value products in biologics manufacturing, a process development based on process understanding and the use of validated process models is imperative. Therefore, a distinct and quantitative validation workflow for liquid-liquid extraction modeling is presented on the example of two complex feed mixtures. Monte-Carlo simulations based on the presented model parameter determination concept result for both examples in prediction accuracy comparable to the experiments and prediction precision within the deviation of the respective experiments. Identification of statistically significant parameters is demonstrated. The presented methodology for model validation will support the implementation of liquid-liquid extraction in the manufacturing of new high value biological products in regulated industries by providing a workflow to derive a Quality-by-Design compatible process model.
Collapse
|
18
|
Accelerating Biologics Manufacturing by Modeling or: Is Approval under the QbD and PAT Approaches Demanded by Authorities Acceptable Without a Digital-Twin? Processes (Basel) 2019. [DOI: 10.3390/pr7020094] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Innovative biologics, including cell therapeutics, virus-like particles, exosomes,recombinant proteins, and peptides, seem likely to substitute monoclonal antibodies as the maintherapeutic entities in manufacturing over the next decades. This molecular variety causes agrowing need for a general change of methods as well as mindset in the process development stage,as there are no platform processes available such as those for monoclonal antibodies. Moreover,market competitiveness demands hyper-intensified processes, including accelerated decisionstoward batch or continuous operation of dedicated modular plant concepts. This indicates gaps inprocess comprehension, when operation windows need to be run at the edges of optimization. Inthis editorial, the authors review and assess potential methods and begin discussing possiblesolutions throughout the workflow, from process development through piloting to manufacturingoperation from their point of view and experience. Especially, the state-of-the-art for modeling inred biotechnology is assessed, clarifying differences and applications of statistical, rigorousphysical-chemical based models as well as cost modeling. “Digital-twins” are described and effortsvs. benefits for new applications exemplified, including the regulation-demanded QbD (quality bydesign) and PAT (process analytical technology) approaches towards digitalization or industry 4.0based on advanced process control strategies. Finally, an analysis of the obstacles and possiblesolutions for any successful and efficient industrialization of innovative methods from processdevelopment, through piloting to manufacturing, results in some recommendations. A centralquestion therefore requires attention: Considering that QbD and PAT have been required byauthorities since 2004, can any biologic manufacturing process be approved by the regulatoryagencies without being modeled by a “digital-twin” as part of the filing documentation?
Collapse
|
19
|
Applications of proteomic methods for CHO host cell protein characterization in biopharmaceutical manufacturing. Curr Opin Biotechnol 2018; 53:144-150. [DOI: 10.1016/j.copbio.2018.01.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/27/2017] [Accepted: 01/04/2018] [Indexed: 12/17/2022]
|
20
|
Kruse T, Kampmann M, Krumbein F, Strube J. Membrane-based separation of aqueous two-phase systems for the purification of monoclonal antibodies. CHEM-ING-TECH 2018. [DOI: 10.1002/cite.201855341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- T. Kruse
- Sartorius Stedim Biotech GmbH; Bioprocessing/ Downstream; August-Spindler-Straße 11 37079 Göttingen Germany
- TU Clausthal; Institut für Thermische Verfahrens- und Prozesstechnik; Leibnizstraße 15 38678 Clausthal-Zellerfeld Germany
| | - M. Kampmann
- Sartorius Stedim Biotech GmbH; Bioprocessing/ Downstream; August-Spindler-Straße 11 37079 Göttingen Germany
| | - F. Krumbein
- Sartorius Stedim Biotech GmbH; Bioprocessing/ Downstream; August-Spindler-Straße 11 37079 Göttingen Germany
| | - J. Strube
- TU Clausthal; Institut für Thermische Verfahrens- und Prozesstechnik; Leibnizstraße 15 38678 Clausthal-Zellerfeld Germany
| |
Collapse
|
21
|
Goey CH, Alhuthali S, Kontoravdi C. Host cell protein removal from biopharmaceutical preparations: Towards the implementation of quality by design. Biotechnol Adv 2018; 36:1223-1237. [DOI: 10.1016/j.biotechadv.2018.03.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 02/12/2018] [Accepted: 03/29/2018] [Indexed: 01/05/2023]
|
22
|
Zobel-Roos S, Stein D, Strube J. Evaluation of Continuous Membrane Chromatography Concepts with an Enhanced Process Simulation Approach. Antibodies (Basel) 2018; 7:E13. [PMID: 31544865 PMCID: PMC6698847 DOI: 10.3390/antib7010013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/20/2018] [Accepted: 02/27/2018] [Indexed: 01/26/2023] Open
Abstract
Modern biopharmaceutical products strive for small-scale, low-cost production. Continuous chromatography has shown to be a promising technology because it assures high-capacity utilization, purity and yield increases, and lower facility footprint. Membrane chromatography is a fully disposable low-cost alternative to bead-based chromatography with minor drawbacks in terms of capacity. Hence, continuous membrane chromatography should have a high potential. The evaluation of continuous processes goes often along with process modeling. Only few experiments with small feed demand need to be conducted to estimate the model parameters. Afterwards, a variety of different process setups and working points can be analyzed in a very short time, making the approach very efficient. Since the available modeling approaches for membrane chromatography modules did not fit the used design, a new modeling approach is shown. This combines the general rate model with an advanced fluid dynamic distribution. Model parameter determination and model validation were done with industrial cell cultures containing Immunoglobulin G (IgG). The validated model was used to evaluate the feasibility of the integrated Counter Current Chromatography (iCCC) concept and the sequential chromatography concept for membrane adsorber modules, starting with a laboratory-type module used for sample preparation. A case study representing a fed-batch reactor with a capacity from 20 to 2000 L was performed. Compared to batch runs, a 71% higher capacity, 48.5% higher productivity, and 38% lower eluent consumption could be achieved.
Collapse
Affiliation(s)
- Steffen Zobel-Roos
- Institute for Separation and Process Technology, Clausthal University of Technology, Leibnizstraße 15, D-38678 Clausthal-Zellerfeld, Germany.
| | - Dominik Stein
- Institute for Separation and Process Technology, Clausthal University of Technology, Leibnizstraße 15, D-38678 Clausthal-Zellerfeld, Germany.
| | - Jochen Strube
- Institute for Separation and Process Technology, Clausthal University of Technology, Leibnizstraße 15, D-38678 Clausthal-Zellerfeld, Germany.
| |
Collapse
|
23
|
Integration of Aqueous Two-Phase Extraction as Cell Harvest and Capture Operation in the Manufacturing Process of Monoclonal Antibodies. Antibodies (Basel) 2017; 6:antib6040021. [PMID: 31548537 PMCID: PMC6698824 DOI: 10.3390/antib6040021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 11/17/2022] Open
Abstract
Substantial improvements have been made to cell culturing processes (e.g., higher product titer) in recent years by raising cell densities and optimizing cultivation time. However, this has been accompanied by an increase in product-related impurities and therefore greater challenges in subsequent clarification and capture operations. Considering the paradigm shift towards the design of continuously operating dedicated plants at smaller scales—with or without disposable technology—for treating smaller patient populations due to new indications or personalized medicine approaches, the rising need for new, innovative strategies for both clarification and capture technology becomes evident. Aqueous two-phase extraction (ATPE) is now considered to be a feasible unit operation, e.g., for the capture of monoclonal antibodies or recombinant proteins. However, most of the published work so far investigates the applicability of ATPE in antibody-manufacturing processes at the lab-scale and for the most part, only during the capture step. This work shows the integration of ATPE as a combined harvest and capture step into a downstream process. Additionally, a model is applied that allows early prediction of settler dimensions with high prediction accuracy. Finally, a reliable process development concept, which guides through the necessary steps, starting from the definition of the separation task to the final stages of integration and scale-up, is presented.
Collapse
|
24
|
Host Cell Proteins in Biologics Manufacturing: The Good, the Bad, and the Ugly. Antibodies (Basel) 2017; 6:antib6030013. [PMID: 31548528 PMCID: PMC6698861 DOI: 10.3390/antib6030013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 01/15/2023] Open
Abstract
Significant progress in the manufacturing of biopharmaceuticals has been made by increasing the overall titers in the USP (upstream processing) titers without raising the cost of the USP. In addition, the development of platform processes led to a higher process robustness. Despite or even due to those achievements, novel challenges are in sight. The higher upstream titers created more complex impurity profiles, both in mass and composition, demanding higher separation capacities and selectivity in downstream processing (DSP). This creates a major shift of costs from USP to DSP. In order to solve this issue, USP and DSP integration approaches can be developed and used for overall process optimization. This study focuses on the characterization and classification of host cell proteins (HCPs) in each unit operation of the DSP (i.e., aqueous two-phase extraction, integrated countercurrent chromatography). The results create a data-driven feedback to the USP, which will serve for media and process optimizations in order to reduce, or even eliminate nascent critical HCPs. This will improve separation efficiency and may lead to a quantitative process understanding. Different HCP species were classified by stringent criteria with regard to DSP separation parameters into “The Good, the Bad, and the Ugly” in terms of pI and MW using 2D-PAGE analysis depending on their positions on the gels. Those spots were identified using LC-MS/MS analysis. HCPs, which are especially difficult to remove and persistent throughout the DSP (i.e., “Bad” or “Ugly”), have to be evaluated by their ability to be separated. In this approach, HCPs, considered “Ugly,” represent proteins with a MW larger than 15 kDa and a pI between 7.30 and 9.30. “Bad” HCPs can likewise be classified using MW (>15 kDa) and pI (4.75–7.30 and 9.30–10.00). HCPs with a MW smaller than 15 kDa and a pI lower than 4.75 and higher than 10.00 are classified as “Good” since their physicochemical properties differ significantly from the product. In order to evaluate this classification scheme, it is of utmost importance to use orthogonal analytical methods such as IEX, HIC, and SEC.
Collapse
|
25
|
Effective strategies for host cell protein clearance in downstream processing of monoclonal antibodies and Fc-fusion proteins. Protein Expr Purif 2017; 134:96-103. [DOI: 10.1016/j.pep.2017.04.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 01/25/2023]
|