1
|
Khodadadi Karimvand S, Cuperlovic-Culf M, Kamen AA, Bolic M. Model-Based Characterization of the Metabolism of Recombinant Adeno-Associated Virus (rAAV) Production via Human Embryonic Kidney (HEK293) Cells. Bioengineering (Basel) 2025; 12:345. [PMID: 40281705 PMCID: PMC12024435 DOI: 10.3390/bioengineering12040345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
In this paper, we present a kinetic-metabolic model describing adeno-associated virus (AAV) production via HEK293 cells that encompasses the main metabolic pathways, namely, glycolysis, tricarboxylic acid cycle (TCA), pyruvate fates, the pentose phosphate pathway, anaplerotic reaction, amino acid metabolism, nucleotides synthesis, biomass synthesis, and the metabolic pathways of protein synthesis of the AAV (capsid and Rep proteins). For the modeling, Michaelis-Menten kinetics was assumed to define the metabolic model. A dataset from bioreactor cultures containing metabolite profiles of adeno-associated virus 6 (AAV6) production via triple transient transfection in a low-cell-density culture, including the concentration profiles of glutamine, glutamic acid, glucose, lactate, and ammonium, was utilized for fitting and computing the model parameters. The model that resulted from the adjusted parameters defined the experimental data well. Subsequently, a Sobol-based global sensitivity analysis procedure was applied to determine the most sensitive parameters in the final model.
Collapse
Affiliation(s)
| | - Miroslava Cuperlovic-Culf
- Digital Technologies Research Center, National Research Council, Ottawa, ON K1A 0R6, Canada;
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Amine A. Kamen
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada;
| | - Miodrag Bolic
- School of Electrical Engineering and Computer Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| |
Collapse
|
2
|
Xu VA, Lee H, Long B, Yuan J, Tang YJ. MAGMA: Microbial and Algal Growth Modeling Application. N Biotechnol 2025; 85:16-22. [PMID: 39603522 DOI: 10.1016/j.nbt.2024.11.004] [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: 08/27/2024] [Revised: 10/29/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Kinetic modeling of biochemical reactions and bioreactor systems can enhance and quantify knowledge gained from cell culture experiments and has many applications in bioprocess design and optimization. The Microbial and Algal Growth Modeling Application (MAGMA) is a user-friendly MATLAB-based software for streamlining the development of kinetic models for various bioreactor systems. This study details the MAGMA workflow by demonstrating the creation of kinetic models with systems of ordinary differential equations (ODEs), model fitting by solving inverse problems, statistical evaluation of model fitting quality, and visual display of simulation results. Two case studies (microalgae growth and Rhodococcus jostii plastic fermentation) have been provided to validate MAGMA applicability. It also includes a proof-of-concept for utilizing OpenAI GPT-4o's graph interpretation capability to automate tabulation of time course culture data from figures/plots in relevant literature, which can be used to calibrate model parameters. MAGMA is open source and compiled with MATLAB Runtime.
Collapse
Affiliation(s)
- Vincent A Xu
- Department of Energy, Environmental, and Chemical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| | - Hakyung Lee
- Department of Energy, Environmental, and Chemical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| | - Bin Long
- Department of Energy, Environmental, and Chemical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| | - Joshua Yuan
- Department of Energy, Environmental, and Chemical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| | - Yinjie J Tang
- Department of Energy, Environmental, and Chemical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
3
|
Kavoni H, Savizi ISP, Lewis NE, Shojaosadati SA. Recent advances in culture medium design for enhanced production of monoclonal antibodies in CHO cells: A comparative study of machine learning and systems biology approaches. Biotechnol Adv 2025; 78:108480. [PMID: 39571767 DOI: 10.1016/j.biotechadv.2024.108480] [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: 06/27/2024] [Revised: 09/24/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
The production of monoclonal antibodies (mAbs) using Chinese Hamster Ovary (CHO) cells has revolutionized the treatment of numerous diseases, solidifying their position as a cornerstone of the biopharmaceutical industry. However, achieving maximum mAb production while upholding strict product quality standards remains a significant hurdle. Optimizing cell culture media emerges as a critical factor in this endeavor, requiring a nuanced understanding of the complex interplay of nutrients, growth factors, and other components that profoundly influence cellular growth, productivity, and product quality. Significant strides have been made in media optimization, including techniques such as media blending, one factor at a time, and statistical design of experiments approaches. The present review provides a comprehensive analysis of the recent advancements in culture media design strategies, focusing on the comparative application of systems biology (SB) and machine learning (ML) approaches. The applications of SB and ML in optimizing CHO cell culture medium and successful examples of their use are summarized. Finally, we highlight the immense potential of integrating SB and ML, emphasizing the development of hybrid models that leverage the strengths of both approaches for robust, efficient, and scalable optimization of mAb production in CHO cells. This review provides a roadmap for researchers and industry professionals to navigate the complex landscape of mAb production optimization, paving the way for developing next-generation CHO cell culture media that drive significant improvements in yield and productivity.
Collapse
Affiliation(s)
- Hossein Kavoni
- Biotechnology Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Iman Shahidi Pour Savizi
- Biotechnology Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Nathan E Lewis
- Department of Bioengineering, University of California, San Diego, CA, USA; Department of Pediatrics, University of California, San Diego, CA, USA
| | - Seyed Abbas Shojaosadati
- Biotechnology Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
4
|
Lu YA, McCann MG, Hu WS, Zhang Q. Multi-cell-line learning for the data-driven construction of mechanistic metabolic models. Biotechnol Bioeng 2024; 121:2833-2847. [PMID: 38831695 DOI: 10.1002/bit.28757] [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: 11/19/2023] [Revised: 04/25/2024] [Accepted: 05/19/2024] [Indexed: 06/05/2024]
Abstract
Mammalian cells are commonly used as hosts in cell culture for biologics production in the pharmaceutical industry. Structured mechanistic models of metabolism have been used to capture complex cellular mechanisms that contribute to varying metabolic shifts in different cell lines. However, little research has focused on the impact of temporal changes in enzyme abundance and activity on the modeling of cell metabolism. In this work, we present a framework for constructing mechanistic models of metabolism that integrate growth-signaling control of enzyme activity and transcript dynamics. The proposed approach is applied to build models for three Chinese hamster ovary (CHO) cell lines using fed-batch culture data and time-series transcript profiles. Leveraging information from the transcriptome data, we develop a parameter estimation approach based on multi-cell-line (MCL) learning, which combines data sets from different cell lines and trains the individual cell-line models jointly to improve model accuracy. The computational results demonstrate the important role of growth signaling and transcript variability in metabolic models as well as the virtue of the MCL approach for constructing cell-line models with a limited amount of data. The resulting models exhibit a high level of accuracy in predicting distinct metabolic behaviors in the different cell lines; these models can potentially be used to accelerate the process and cell-line development for the biomanufacturing of new protein therapeutics.
Collapse
Affiliation(s)
- Yen-An Lu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Meghan G McCann
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Qi Zhang
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
5
|
Weggen JT, Bean R, Hui K, Wendeler M, Hubbuch J. Kinetic models towards an enhanced understanding of diverse ADC conjugation reactions. Front Bioeng Biotechnol 2024; 12:1403644. [PMID: 39070164 PMCID: PMC11274341 DOI: 10.3389/fbioe.2024.1403644] [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: 03/19/2024] [Accepted: 06/07/2024] [Indexed: 07/30/2024] Open
Abstract
The conjugation reaction is the central step in the manufacturing process of antibody-drug conjugates (ADCs). This reaction generates a heterogeneous and complex mixture of differently conjugated sub-species depending on the chosen conjugation chemistry. The parametrization of the conjugation reaction through mechanistic kinetic models offers a chance to enhance valuable reaction knowledge and ensure process robustness. This study introduces a versatile modeling framework for the conjugation reaction of cysteine-conjugated ADC modalities-site-specific and interchain disulfide conjugation. Various conjugation kinetics involving different maleimide-functionalized payloads were performed, while controlled gradual payload feeding was employed to decelerate the conjugation, facilitating a more detailed investigation of the reaction mechanism. The kinetic data were analyzed with a reducing reversed phase (RP) chromatography method, that can readily be implemented for the accurate characterization of ADCs with diverse drug-to-antibody ratios, providing the conjugation trajectories of the single chains of the monoclonal antibody (mAb). Possible kinetic models for the conjugation mechanism were then developed and selected based on multiple criteria. When calibrating the established model to kinetics involving different payloads, conjugation rates were determined to be payload-specific. Further conclusions regarding the kinetic comparability across the two modalities could also be derived. One calibrated model was used for an exemplary in silico screening of the initial concentrations offering valuable insights for profound understanding of the conjugation process in ADC development.
Collapse
Affiliation(s)
- Jan Tobias Weggen
- Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Ryan Bean
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Kimberly Hui
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Michaela Wendeler
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Jürgen Hubbuch
- Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
6
|
Ma C, Gurkan-Cavusoglu E. A comprehensive review of computational cell cycle models in guiding cancer treatment strategies. NPJ Syst Biol Appl 2024; 10:71. [PMID: 38969664 PMCID: PMC11226463 DOI: 10.1038/s41540-024-00397-7] [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: 01/26/2024] [Accepted: 06/24/2024] [Indexed: 07/07/2024] Open
Abstract
This article reviews the current knowledge and recent advancements in computational modeling of the cell cycle. It offers a comparative analysis of various modeling paradigms, highlighting their unique strengths, limitations, and applications. Specifically, the article compares deterministic and stochastic models, single-cell versus population models, and mechanistic versus abstract models. This detailed analysis helps determine the most suitable modeling framework for various research needs. Additionally, the discussion extends to the utilization of these computational models to illuminate cell cycle dynamics, with a particular focus on cell cycle viability, crosstalk with signaling pathways, tumor microenvironment, DNA replication, and repair mechanisms, underscoring their critical roles in tumor progression and the optimization of cancer therapies. By applying these models to crucial aspects of cancer therapy planning for better outcomes, including drug efficacy quantification, drug discovery, drug resistance analysis, and dose optimization, the review highlights the significant potential of computational insights in enhancing the precision and effectiveness of cancer treatments. This emphasis on the intricate relationship between computational modeling and therapeutic strategy development underscores the pivotal role of advanced modeling techniques in navigating the complexities of cell cycle dynamics and their implications for cancer therapy.
Collapse
Affiliation(s)
- Chenhui Ma
- Department of Electrical, Computer and Systems Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - Evren Gurkan-Cavusoglu
- Department of Electrical, Computer and Systems Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
7
|
Buick E, Mead A, Alhubaysh A, Bou Assi P, Das P, Dayus J, Turner M, Kowalski L, Murray J, Renshaw D, Farnaud S. CellShip: An Ambient Temperature Transport and Short-Term Storage Medium for Mammalian Cell Cultures. Biopreserv Biobank 2024; 22:275-285. [PMID: 38150708 DOI: 10.1089/bio.2023.0100] [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: 12/29/2023] Open
Abstract
Cell culture is a critical platform for numerous research and industrial processes. However, methods for transporting cells are largely limited to cryopreservation, which is logistically challenging, requires the use of potentially cytotoxic cryopreservatives, and can result in poor cell recovery. Development of a transport media that can be used at ambient temperatures would alleviate these issues. In this study, we describe a novel transportation medium for mammalian cells. Five commonly used cell lines, (HEK293, CHO, HepG2, K562, and Jurkat) were successfully shipped and stored for a minimum of 72 hours and up to 96 hours at ambient temperature, after which, cells were recovered into standard culture conditions. Viability (%) and cell numbers, were examined, before, following the transport/storage period and following the recovery period. In all experiments, cell numbers returned to pretransport/storage concentration within 24-48 hours recovery. Imaging data indicated that HepG2 cells were fully adherent and had established typical growth morphology following 48 hours recovery, which was not seen in cells recovered from cryopreservation. Following recovery, Jurkat cells that had been subjected to a 96 hours transport/storage period, demonstrated a 1.93-fold increase compared with the starting cell number with >95% cell viability. We conclude that CellShip® may represent a viable method for the transportation of mammalian cells for multiple downstream applications in the Life Sciences research sector.
Collapse
Affiliation(s)
- Emma Buick
- Life Science Production, Bedford, United Kingdom
- Center of Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Andrew Mead
- Comparative Biomedical Sciences, The Royal Veterinary College (RVC), London, United Kingdom
| | | | | | - Parijat Das
- Life Science Production, Bedford, United Kingdom
| | - James Dayus
- Center of Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom
- Faculty of Health and Life Sciences, School of Life Sciences, Coventry University, Coventry, United Kingdom
| | - Mark Turner
- Center of Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Lukasz Kowalski
- Life Science Production, Bedford, United Kingdom
- Center of Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Jenny Murray
- Life Science Production, Bedford, United Kingdom
| | - Derek Renshaw
- Center of Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Sebastien Farnaud
- Center of Sport, Exercise and Life Sciences, Coventry University, Coventry, United Kingdom
| |
Collapse
|
8
|
González-Hernández Y, Perré P. Building blocks needed for mechanistic modeling of bioprocesses: A critical review based on protein production by CHO cells. Metab Eng Commun 2024; 18:e00232. [PMID: 38501051 PMCID: PMC10945193 DOI: 10.1016/j.mec.2024.e00232] [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: 10/25/2023] [Revised: 02/12/2024] [Accepted: 02/23/2024] [Indexed: 03/20/2024] Open
Abstract
This paper reviews the key building blocks needed to develop a mechanistic model for use as an operational production tool. The Chinese Hamster Ovary (CHO) cell, one of the most widely used hosts for antibody production in the pharmaceutical industry, is considered as a case study. CHO cell metabolism is characterized by two main phases, exponential growth followed by a stationary phase with strong protein production. This process presents an appropriate degree of complexity to outline the modeling strategy. The paper is organized into four main steps: (1) CHO systems and data collection; (2) metabolic analysis; (3) formulation of the mathematical model; and finally, (4) numerical solution, calibration, and validation. The overall approach can build a predictive model of target variables. According to the literature, one of the main current modeling challenges lies in understanding and predicting the spontaneous metabolic shift. Possible candidates for the trigger of the metabolic shift include the concentration of lactate and carbon dioxide. In our opinion, ammonium, which is also an inhibiting product, should be further investigated. Finally, the expected progress in the emerging field of hybrid modeling, which combines the best of mechanistic modeling and machine learning, is presented as a fascinating breakthrough. Note that the modeling strategy discussed here is a general framework that can be applied to any bioprocess.
Collapse
Affiliation(s)
- Yusmel González-Hernández
- Université Paris-Saclay, CentraleSupélec, Laboratoire de Génie des Procédés et Matériaux, Centre Européen de Biotechnologie et de Bioéconomie (CEBB), 3 Rue des Rouges Terres, 51110, Pomacle, France
| | - Patrick Perré
- Université Paris-Saclay, CentraleSupélec, Laboratoire de Génie des Procédés et Matériaux, Centre Européen de Biotechnologie et de Bioéconomie (CEBB), 3 Rue des Rouges Terres, 51110, Pomacle, France
| |
Collapse
|
9
|
Kim YS, Lee JS, Jeong MY, Jang JW, Kim MS. Recombinant human fibroblast growth factor 7 obtained from stable Chinese hamster ovary cells enhances wound healing. Biotechnol J 2024; 19:e2300596. [PMID: 38719591 DOI: 10.1002/biot.202300596] [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: 11/01/2023] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 05/14/2024]
Abstract
Although fibroblast growth factor 7 (FGF7) is known to promote wound healing, its mass production poses several challenges and very few studies have assessed the feasibility of producing FGF7 in cell lines such as Chinese hamster ovary (CHO) cells. Therefore, this study sought to produce recombinant FGF7 in large quantities and evaluate its wound healing effect. To this end, the FGF7 gene was transfected into CHO cells and FGF7 production was optimized. The wound healing efficacy of N-glycosylated FGF7 was evaluated in animals on days 7 and 14 post-treatment using collagen patches (CPs), FGF7-only, and CP with FGF7 (CP+FGF7), whereas an untreated group was used as the control. Wound healing was most effective in the CP+FGF7 group. Particularly, on day 7 post-exposure, the CP+FGF7 and FGF7-only groups exhibited the highest expression of hydroxyproline, fibroblast growth factor, vascular endothelial growth factor, and transforming growth factor. Epidermalization in H&E staining showed the same order of healing as hydroxyproline content. Additionally, the CP+FGF7 and FGF7-only group exhibited more notable blood vessel formation on days 7 and 14. In conclusion, the prepared FGF7 was effective in promoting wound healing and CHO cells can be a reliable platform for the mass production of FGF7.
Collapse
Affiliation(s)
- Young Sik Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- The Institute of Biomaterial and Medical Engineering, Cellumed Co., Ltd., Seoul, Sourh Korea
| | - Jung Soo Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- The Institute of Biomaterial and Medical Engineering, Cellumed Co., Ltd., Seoul, Sourh Korea
| | - Mi Yeong Jeong
- The Institute of Biomaterial and Medical Engineering, Cellumed Co., Ltd., Seoul, Sourh Korea
| | - Ju Woong Jang
- The Institute of Biomaterial and Medical Engineering, Cellumed Co., Ltd., Seoul, Sourh Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
10
|
Wang K, Xie W, Harcum SW. Metabolic regulatory network kinetic modeling with multiple isotopic tracers for iPSCs. Biotechnol Bioeng 2024; 121:1336-1354. [PMID: 38037741 DOI: 10.1002/bit.28609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/26/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023]
Abstract
The rapidly expanding market for regenerative medicines and cell therapies highlights the need to advance the understanding of cellular metabolisms and improve the prediction of cultivation production process for human induced pluripotent stem cells (iPSCs). In this paper, a metabolic kinetic model was developed to characterize the underlying mechanisms of iPSC culture process, which can predict cell response to environmental perturbation and support process control. This model focuses on the central carbon metabolic network, including glycolysis, pentose phosphate pathway, tricarboxylic acid cycle, and amino acid metabolism, which plays a crucial role to support iPSC proliferation. Heterogeneous measures of extracellular metabolites and multiple isotopic tracers collected under multiple conditions were used to learn metabolic regulatory mechanisms. Systematic cross-validation confirmed the model's performance in terms of providing reliable predictions on cellular metabolism and culture process dynamics under various culture conditions. Thus, the developed mechanistic kinetic model can support process control strategies to strategically select optimal cell culture conditions at different times, ensure cell product functionality, and facilitate large-scale manufacturing of regenerative medicines and cell therapies.
Collapse
Affiliation(s)
- Keqi Wang
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Wei Xie
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Sarah W Harcum
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
11
|
Xing Z, Nguyen TB, Kanai-Bai G, Yamano-Adachi N, Omasa T. Construction of a novel kinetic model for the production process of a CVA6 VLP vaccine in CHO cells. Cytotechnology 2024; 76:69-83. [PMID: 38304624 PMCID: PMC10828271 DOI: 10.1007/s10616-023-00598-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/22/2023] [Indexed: 02/03/2024] Open
Abstract
Bioprocess development benefits from kinetic models in many aspects, including scale-up, optimization, and process understanding. However, current models are unable to simulate the production process of a coxsackievirus A6 (CVA6) virus-like particle (VLP) vaccine using Chinese hamster ovary cell culture. In this study, a novel kinetic model was constructed, correlating (1) cell growth, death, and lysis kinetics, (2) metabolism of major metabolites, and (3) CVA6 VLP production. To construct the model, two batches of a laboratory-scale 2 L bioreactor cell culture were prepared and various pH shift strategies were applied to examine the effect of pH shift. The proposed model described the experimental data under various conditions with high accuracy and quantified the effect of pH shift. Next, cell culture performance with various pH shift timings was predicted by the calibrated model. A trade-off relationship was found between product yield and quality. Consequently, multiple objective optimization was performed by integrating desirability methodology with model simulation. Finally, the optimal operating conditions that balanced product yield and quality were predicted. In general, the proposed model improved the process understanding and enabled in silico process development of a CVA6 VLP vaccine. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-023-00598-8.
Collapse
Affiliation(s)
- Zhou Xing
- Graduate School of Engineering, Osaka University, U1E801, 2-1Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Thao Bich Nguyen
- Graduate School of Engineering, Osaka University, U1E801, 2-1Yamadaoka, Suita, Osaka 565-0871 Japan
- Present Address: Tsukuba Research Laboratories, Eisai Co. Ltd, 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635 Japan
| | - Guirong Kanai-Bai
- Graduate School of Engineering, Osaka University, U1E801, 2-1Yamadaoka, Suita, Osaka 565-0871 Japan
- Institute for Open and Transdisciplinary Research Initiatives, U1E801, 2-1 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Noriko Yamano-Adachi
- Graduate School of Engineering, Osaka University, U1E801, 2-1Yamadaoka, Suita, Osaka 565-0871 Japan
- Institute for Open and Transdisciplinary Research Initiatives, U1E801, 2-1 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Takeshi Omasa
- Graduate School of Engineering, Osaka University, U1E801, 2-1Yamadaoka, Suita, Osaka 565-0871 Japan
- Institute for Open and Transdisciplinary Research Initiatives, U1E801, 2-1 Yamadaoka, Suita, Osaka 565-0871 Japan
| |
Collapse
|
12
|
Iglesias CF, Bolic M. How Not to Make the Joint Extended Kalman Filter Fail with Unstructured Mechanistic Models. SENSORS (BASEL, SWITZERLAND) 2024; 24:653. [PMID: 38276345 PMCID: PMC11154378 DOI: 10.3390/s24020653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/22/2023] [Accepted: 01/06/2024] [Indexed: 01/27/2024]
Abstract
The unstructured mechanistic model (UMM) allows for modeling the macro-scale of a phenomenon without known mechanisms. This is extremely useful in biomanufacturing because using the UMM for the joint estimation of states and parameters with an extended Kalman filter (JEKF) can enable the real-time monitoring of bioprocesses with unknown mechanisms. However, the UMM commonly used in biomanufacturing contains ordinary differential equations (ODEs) with unshared parameters, weak variables, and weak terms. When such a UMM is coupled with an initial state error covariance matrix P(t=0) and a process error covariance matrix Q with uncorrelated elements, along with just one measured state variable, the joint extended Kalman filter (JEKF) fails to estimate the unshared parameters and state simultaneously. This is because the Kalman gain corresponding to the unshared parameter remains constant and equal to zero. In this work, we formally describe this failure case, present the proof of JEKF failure, and propose an approach called SANTO to side-step this failure case. The SANTO approach consists of adding a quantity to the state error covariance between the measured state variable and unshared parameter in the initial P(t = 0) of the matrix Ricatti differential equation to compute the predicted error covariance matrix of the state and prevent the Kalman gain from being zero. Our empirical evaluations using synthetic and real datasets reveal significant improvements: SANTO achieved a reduction in root-mean-square percentage error (RMSPE) of up to approximately 17% compared to the classical JEKF, indicating a substantial enhancement in estimation accuracy.
Collapse
Affiliation(s)
- Cristovão Freitas Iglesias
- School of Electrical Engineering and Computer Science (EECS), University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Miodrag Bolic
- School of Electrical Engineering and Computer Science (EECS), University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
13
|
Zheng H, Harcum SW, Pei J, Xie W. Stochastic biological system-of-systems modelling for iPSC culture. Commun Biol 2024; 7:39. [PMID: 38191636 PMCID: PMC10774284 DOI: 10.1038/s42003-023-05653-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/30/2023] [Indexed: 01/10/2024] Open
Abstract
Large-scale manufacturing of induced pluripotent stem cells (iPSCs) is essential for cell therapies and regenerative medicines. Yet, iPSCs form large cell aggregates in suspension bioreactors, resulting in insufficient nutrient supply and extra metabolic waste build-up for the cells located at the core. Since subtle changes in micro-environment can lead to a heterogeneous cell population, a novel Biological System-of-Systems (Bio-SoS) framework is proposed to model cell-to-cell interactions, spatial and metabolic heterogeneity, and cell response to micro-environmental variation. Building on stochastic metabolic reaction network, aggregation kinetics, and reaction-diffusion mechanisms, the Bio-SoS model characterizes causal interdependencies at individual cell, aggregate, and cell population levels. It has a modular design that enables data integration and improves predictions for different monolayer and aggregate culture processes. In addition, a variance decomposition analysis is derived to quantify the impact of factors (i.e., aggregate size) on cell product health and quality heterogeneity.
Collapse
Affiliation(s)
- Hua Zheng
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | | | - Jinxiang Pei
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Wei Xie
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
14
|
Pogodaev A, Hernández Rodríguez T, Li M, García Münzer DG. Modeling of bioprocess pre-stages for optimization of perfusion profiles and increased process understanding. Biotechnol Bioeng 2024; 121:228-237. [PMID: 37902718 DOI: 10.1002/bit.28576] [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: 03/28/2023] [Revised: 08/18/2023] [Accepted: 10/14/2023] [Indexed: 10/31/2023]
Abstract
Improving bioprocess efficiency is important to reduce the current costs of biologics on the market, bring them faster to the market, and to improve the environmental footprint. The process intensification efforts were historically focused on the main stage, while intensification of pre-stages has started to gain attention only in the past decade. Performing bioprocess pre-stages in the perfusion mode is one of the most efficient options to achieve higher viable cell densities over traditional batch methods. While the perfusion-mode operation allows to reach higher viable cell densities, it also consumes large amount of medium, making it cost-intensive. The change of perfusion rate during a process (perfusion profile) determines how much medium is consumed, thereby running a process in optimal conditions is key to reduce medium consumption. However, the selection of the perfusion profile is often made empirically, without full understanding of bioprocess dynamics. This fact is hindering potential process improvements and means for cost reduction. In this study, we propose a process modeling approach to identify the optimal perfusion profile during bioprocess pre-stages. The developed process model was used internally during process development. We could reduce perfused medium volume by 25%-45% (project-dependent), while keeping the difference in the final cell within 5%-10% compared to the original settings. Additionally, the model helps to reduce the experimental workload by 30%-70% and to predict an optimal perfusion profile when process conditions need to be changed (e.g., higher seeding density, change of operating mode from batch to perfusion, etc.). This study demonstrates the potential of process modeling as a powerful tool for optimizing bioprocess pre-stages and thereby guiding process development, improving overall bioprocess efficiency, and reducing operational costs, while strongly reducing the need for wet-lab experiments.
Collapse
Affiliation(s)
| | | | - Mengyao Li
- Novartis Technical Research & Development, Basel, Switzerland
| | | |
Collapse
|
15
|
Reddy JV, Raudenbush K, Papoutsakis ET, Ierapetritou M. Cell-culture process optimization via model-based predictions of metabolism and protein glycosylation. Biotechnol Adv 2023; 67:108179. [PMID: 37257729 DOI: 10.1016/j.biotechadv.2023.108179] [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: 11/27/2022] [Revised: 05/18/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
In order to meet the rising demand for biologics and become competitive on the developing biosimilar market, there is a need for process intensification of biomanufacturing processes. Process development of biologics has historically relied on extensive experimentation to develop and optimize biopharmaceutical manufacturing. Experimentation to optimize media formulations, feeding schedules, bioreactor operations and bioreactor scale up is expensive, labor intensive and time consuming. Mathematical modeling frameworks have the potential to enable process intensification while reducing the experimental burden. This review focuses on mathematical modeling of cellular metabolism and N-linked glycosylation as applied to upstream manufacturing of biologics. We review developments in the field of modeling cellular metabolism of mammalian cells using kinetic and stoichiometric modeling frameworks along with their applications to simulate, optimize and improve mechanistic understanding of the process. Interest in modeling N-linked glycosylation has led to the creation of various types of parametric and non-parametric models. Most published studies on mammalian cell metabolism have performed experiments in shake flasks where the pH and dissolved oxygen cannot be controlled. Efforts to understand and model the effect of bioreactor-specific parameters such as pH, dissolved oxygen, temperature, and bioreactor heterogeneity are critically reviewed. Most modeling efforts have focused on the Chinese Hamster Ovary (CHO) cells, which are most commonly used to produce monoclonal antibodies (mAbs). However, these modeling approaches can be generalized and applied to any mammalian cell-based manufacturing platform. Current and potential future applications of these models for Vero cell-based vaccine manufacturing, CAR-T cell therapies, and viral vector manufacturing are also discussed. We offer specific recommendations for improving the applicability of these models to industrially relevant processes.
Collapse
Affiliation(s)
- Jayanth Venkatarama Reddy
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA
| | - Katherine Raudenbush
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA; Delaware Biotechnology Institute, Department of Biological Sciences, University of Delaware, USA.
| | - Marianthi Ierapetritou
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA.
| |
Collapse
|
16
|
Masson HO, Karottki KJLC, Tat J, Hefzi H, Lewis NE. From observational to actionable: rethinking omics in biologics production. Trends Biotechnol 2023; 41:1127-1138. [PMID: 37062598 PMCID: PMC10524802 DOI: 10.1016/j.tibtech.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 04/18/2023]
Abstract
As the era of omics continues to expand with increasing ubiquity and success in both academia and industry, omics-based experiments are becoming commonplace in industrial biotechnology, including efforts to develop novel solutions in bioprocess optimization and cell line development. Omic technologies provide particularly valuable 'observational' insights for discovery science, especially in academic research and industrial R&D; however, biomanufacturing requires a different paradigm to unlock 'actionable' insights from omics. Here, we argue the value of omic experiments in biotechnology can be maximized with deliberate selection of omic approaches and forethought about analysis techniques. We describe important considerations when designing and implementing omic-based experiments and discuss how systems biology analysis strategies can enhance efforts to obtain actionable insights in mammalian-based biologics production.
Collapse
Affiliation(s)
- Helen O Masson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | - Jasmine Tat
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Amgen Inc., Thousand Oaks, CA, USA
| | | | - Nathan E Lewis
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
17
|
Rapid Identification of Chinese Hamster Ovary Cell Apoptosis and Its Potential Role in Process Robustness Assessment. Bioengineering (Basel) 2023; 10:bioengineering10030357. [PMID: 36978748 PMCID: PMC10045091 DOI: 10.3390/bioengineering10030357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Currently, the assessment of process robustness is often time-consuming, labor-intensive, and material-intensive using process characterization studies. Therefore, a simple and time-saving method is highly needed for the biopharmaceutical industry. Apoptosis is responsible for 80% of Chinese hamster ovary (CHO) cell deaths and affects the robustness of the cell culture process. This study’s results showed that a more robust process can support cells to tolerate apoptosis for a longer time, suggesting that the robustness of the process could be judged by the ability of cells to resist apoptosis. Therefore, it is necessary to establish a rapid method to detect the apoptosis of CHO cells. In trying to establish a new method for detecting apoptosis in large-scale cell cultures, glucose withdrawal was studied, and the results showed that CHO cells began to apoptose after glucose was consumed. Then, the concentration of extracellular potassium increased, and a prolongation of apoptosis time was observed. Further study results showed that the process with poor robustness was associated with a higher proportion of apoptosis and extracellular potassium concentration, so potassium could be used as a biochemical index of apoptosis. The strategy we present may be used to expedite the assessment of process robustness to obtain a robust cell culture process for other biologics.
Collapse
|
18
|
Canova CT, Inguva PK, Braatz RD. Mechanistic modeling of viral particle production. Biotechnol Bioeng 2023; 120:629-641. [PMID: 36461898 DOI: 10.1002/bit.28296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022]
Abstract
Viral systems such as wild-type viruses, viral vectors, and virus-like particles are essential components of modern biotechnology and medicine. Despite their importance, the commercial-scale production of viral systems remains highly inefficient for multiple reasons. Computational strategies are a promising avenue for improving process development, optimization, and control, but require a mathematical description of the system. This article reviews mechanistic modeling strategies for the production of viral particles, both at the cellular and bioreactor scales. In many cases, techniques and models from adjacent fields such as epidemiology and wild-type viral infection kinetics can be adapted to construct a suitable process model. These process models can then be employed for various purposes such as in-silico testing of novel process operating strategies and/or advanced process control.
Collapse
Affiliation(s)
- Christopher T Canova
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Pavan K Inguva
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard D Braatz
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
19
|
Iglesias CF, Ristovski M, Bolic M, Cuperlovic-Culf M. rAAV Manufacturing: The Challenges of Soft Sensing during Upstream Processing. Bioengineering (Basel) 2023; 10:bioengineering10020229. [PMID: 36829723 PMCID: PMC9951952 DOI: 10.3390/bioengineering10020229] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) is the most effective viral vector technology for directly translating the genomic revolution into medicinal therapies. However, the manufacturing of rAAV viral vectors remains challenging in the upstream processing with low rAAV yield in large-scale production and high cost, limiting the generalization of rAAV-based treatments. This situation can be improved by real-time monitoring of critical process parameters (CPP) that affect critical quality attributes (CQA). To achieve this aim, soft sensing combined with predictive modeling is an important strategy that can be used for optimizing the upstream process of rAAV production by monitoring critical process variables in real time. However, the development of soft sensors for rAAV production as a fast and low-cost monitoring approach is not an easy task. This review article describes four challenges and critically discusses the possible solutions that can enable the application of soft sensors for rAAV production monitoring. The challenges from a data scientist's perspective are (i) a predictor variable (soft-sensor inputs) set without AAV viral titer, (ii) multi-step forecasting, (iii) multiple process phases, and (iv) soft-sensor development composed of the mechanistic model.
Collapse
Affiliation(s)
| | - Milica Ristovski
- Faculty of Engineering, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Miodrag Bolic
- Faculty of Engineering, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Miroslava Cuperlovic-Culf
- Digital Technologies Research Center, National Research Council, Ottawa, ON K1A 0R6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Correspondence:
| |
Collapse
|
20
|
Mowbray MR, Wu C, Rogers AW, Rio-Chanona EAD, Zhang D. A reinforcement learning-based hybrid modeling framework for bioprocess kinetics identification. Biotechnol Bioeng 2023; 120:154-168. [PMID: 36225098 PMCID: PMC10092184 DOI: 10.1002/bit.28262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 07/18/2022] [Accepted: 10/09/2022] [Indexed: 11/09/2022]
Abstract
Constructing predictive models to simulate complex bioprocess dynamics, particularly time-varying (i.e., parameters varying over time) and history-dependent (i.e., current kinetics dependent on historical culture conditions) behavior, has been a longstanding research challenge. Current advances in hybrid modeling offer a solution to this by integrating kinetic models with data-driven techniques. This article proposes a novel two-step framework: first (i) speculate and combine several possible kinetic model structures sourced from process and phenomenological knowledge, then (ii) identify the most likely kinetic model structure and its parameter values using model-free Reinforcement Learning (RL). Specifically, Step 1 collates feasible history-dependent model structures, then Step 2 uses RL to simultaneously identify the correct model structure and the time-varying parameter trajectories. To demonstrate the performance of this framework, a range of in-silico case studies were carried out. The results show that the proposed framework can efficiently construct high-fidelity models to quantify both time-varying and history-dependent kinetic behaviors while minimizing the risks of over-parametrization and over-fitting. Finally, the primary advantages of the proposed framework and its limitation were thoroughly discussed in comparison to other existing hybrid modeling and model structure identification techniques, highlighting the potential of this framework for general bioprocess modeling.
Collapse
Affiliation(s)
- Max R Mowbray
- Department of Chemical Engineering, Centre for Process Integration, University of Manchester, Manchester, UK
| | - Chufan Wu
- Department of Chemical Engineering, Centre for Process Integration, University of Manchester, Manchester, UK
| | - Alexander W Rogers
- Department of Chemical Engineering, Centre for Process Integration, University of Manchester, Manchester, UK
| | | | - Dongda Zhang
- Department of Chemical Engineering, Centre for Process Integration, University of Manchester, Manchester, UK
| |
Collapse
|
21
|
Doyle K, Tsopanoglou A, Fejér A, Glennon B, del Val IJ. Automated assembly of hybrid dynamic models for CHO cell culture processes. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
22
|
Yeo HC, Selvarajoo K. Machine learning alternative to systems biology should not solely depend on data. Brief Bioinform 2022; 23:6731718. [PMID: 36184188 PMCID: PMC9677488 DOI: 10.1093/bib/bbac436] [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: 05/06/2022] [Revised: 08/24/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022] Open
Abstract
In recent years, artificial intelligence (AI)/machine learning has emerged as a plausible alternative to systems biology for the elucidation of biological phenomena and in attaining specified design objective in synthetic biology. Although considered highly disruptive with numerous notable successes so far, we seek to bring attention to both the fundamental and practical pitfalls of their usage, especially in illuminating emergent behaviors from chaotic or stochastic systems in biology. Without deliberating on their suitability and the required data qualities and pre-processing approaches beforehand, the research and development community could experience similar 'AI winters' that had plagued other fields. Instead, we anticipate the integration or combination of the two approaches, where appropriate, moving forward.
Collapse
Affiliation(s)
- Hock Chuan Yeo
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore
| | | |
Collapse
|
23
|
Hartmann FSF, Udugama IA, Seibold GM, Sugiyama H, Gernaey KV. Digital models in biotechnology: Towards multi-scale integration and implementation. Biotechnol Adv 2022; 60:108015. [PMID: 35781047 DOI: 10.1016/j.biotechadv.2022.108015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/03/2022] [Accepted: 06/27/2022] [Indexed: 12/28/2022]
Abstract
Industrial biotechnology encompasses a large area of multi-scale and multi-disciplinary research activities. With the recent megatrend of digitalization sweeping across all industries, there is an increased focus in the biotechnology industry on developing, integrating and applying digital models to improve all aspects of industrial biotechnology. Given the rapid development of this field, we systematically classify the state-of-art modelling concepts applied at different scales in industrial biotechnology and critically discuss their current usage, advantages and limitations. Further, we critically analyzed current strategies to couple cell models with computational fluid dynamics to study the performance of industrial microorganisms in large-scale bioprocesses, which is of crucial importance for the bio-based production industries. One of the most challenging aspects in this context is gathering intracellular data under industrially relevant conditions. Towards comprehensive models, we discuss how different scale-down concepts combined with appropriate analytical tools can capture intracellular states of single cells. We finally illustrated how the efforts could be used to develop digitals models suitable for both cell factory design and process optimization at industrial scales in the future.
Collapse
Affiliation(s)
- Fabian S F Hartmann
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Building 223, 2800 Kgs. Lyngby, Denmark
| | - Isuru A Udugama
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-8656 Tokyo, Japan; Department of Chemical and Biochemical Engineering, Technical University of Denmark, Søltofts Plads, Building 228 A, 2800 Kgs. Lyngby, Denmark.
| | - Gerd M Seibold
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Building 223, 2800 Kgs. Lyngby, Denmark
| | - Hirokazu Sugiyama
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-8656 Tokyo, Japan
| | - Krist V Gernaey
- Department of Chemical and Biochemical Engineering, Technical University of Denmark, Søltofts Plads, Building 228 A, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
24
|
Wang Z, Wang C, Chen G. Kinetic modeling: A tool for temperature shift and feeding optimization in cell culture process development. Protein Expr Purif 2022; 198:106130. [PMID: 35691496 DOI: 10.1016/j.pep.2022.106130] [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: 03/08/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022]
Abstract
Mammalian cells have dominated the biopharmaceutical industry for biotherapeutic protein production and tremendous efforts have been devoted to enhancing productivity during the cell culture process development. However, determining the optimal process conditions is still a huge challenge. Constrained by the limited resources and timeline, usually it is impossible to fully explore the optimal range of all process parameters (temperature, pH, dissolved oxygen, basal and feeding medium, additives, etc.). Kinetic modeling, which finds out the global optimum by systematically screening all potential conditions for cell culture process, provides a solution to this dilemma. However, studies on optimizing temperature shift and feeding strategies simultaneously using this approach have not been reported. In this study, we built up a kinetic model of fed-batch culture process for simultaneous optimization of temperature shift and feeding strategies. The fitting results showed high accuracy and demonstrated that the kinetic model can be used to describe the mammalian cell culture performance. In addition, five more fed-batch experiments were conducted to test this model's predicting power on different temperature shift and feeding strategies. It turned out that the predicted data matched well with experimental ones on viable cell density (VCD), metabolites, and titer for the entire culture duration and allowed selecting the same best condition with the experimental results. Therefore, adopting this approach can potentially reduce the number of experiments required for culture process optimization.
Collapse
Affiliation(s)
- Zheyu Wang
- Technology and Process Development (TPD), WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Caixia Wang
- Technology and Process Development (TPD), WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Gong Chen
- Technology and Process Development (TPD), WuXi Biologics, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China.
| |
Collapse
|
25
|
Joiner J, Huang Z, McHugh K, Stebbins M, Aron K, Borys M, Khetan A. Process modeling of recombinant adeno-associated virus production in HEK293 cells. Curr Opin Chem Eng 2022. [DOI: 10.1016/j.coche.2022.100823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
26
|
Hirono K, A. Udugama I, Hayashi Y, Kino-oka M, Sugiyama H. A Dynamic and Probabilistic Design Space Determination Method for Mesenchymal Stem Cell Cultivation Processes. Ind Eng Chem Res 2022. [DOI: 10.1021/acs.iecr.2c00374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Keita Hirono
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Isuru A. Udugama
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yusuke Hayashi
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Masahiro Kino-oka
- Department of Biotechnology, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hirokazu Sugiyama
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
27
|
Pajčin I, Knežić T, Savic Azoulay I, Vlajkov V, Djisalov M, Janjušević L, Grahovac J, Gadjanski I. Bioengineering Outlook on Cultivated Meat Production. MICROMACHINES 2022; 13:402. [PMID: 35334693 PMCID: PMC8950996 DOI: 10.3390/mi13030402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023]
Abstract
Cultured meat (also referred to as cultivated meat or cell-based meat)-CM-is fabricated through the process of cellular agriculture (CA), which entails application of bioengineering, i.e., tissue engineering (TE) principles to the production of food. The main TE principles include usage of cells, grown in a controlled environment provided by bioreactors and cultivation media supplemented with growth factors and other needed nutrients and signaling molecules, and seeded onto the immobilization elements-microcarriers and scaffolds that provide the adhesion surfaces necessary for anchor-dependent cells and offer 3D organization for multiple cell types. Theoretically, many solutions from regenerative medicine and biomedical engineering can be applied in CM-TE, i.e., CA. However, in practice, there are a number of specificities regarding fabrication of a CM product that needs to fulfill not only the majority of functional criteria of muscle and fat TE, but also has to possess the sensory and nutritional qualities of a traditional food component, i.e., the meat it aims to replace. This is the reason that bioengineering aimed at CM production needs to be regarded as a specific scientific discipline of a multidisciplinary nature, integrating principles from biomedical engineering as well as from food manufacturing, design and development, i.e., food engineering. An important requirement is also the need to use as little as possible of animal-derived components in the whole CM bioprocess. In this review, we aim to present the current knowledge on different bioengineering aspects, pertinent to different current scientific disciplines but all relevant for CM engineering, relevant for muscle TE, including different cell sources, bioreactor types, media requirements, bioprocess monitoring and kinetics and their modifications for use in CA, all in view of their potential for efficient CM bioprocess scale-up. We believe such a review will offer a good overview of different bioengineering strategies for CM production and will be useful to a range of interested stakeholders, from students just entering the CA field to experienced researchers looking for the latest innovations in the field.
Collapse
Affiliation(s)
- Ivana Pajčin
- Department of Biotechnology and Pharmaceutical Engineering, Faculty of Technology Novi Sad, University of Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia; (I.P.); (V.V.); (J.G.)
| | - Teodora Knežić
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (M.D.); (L.J.)
| | - Ivana Savic Azoulay
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel;
| | - Vanja Vlajkov
- Department of Biotechnology and Pharmaceutical Engineering, Faculty of Technology Novi Sad, University of Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia; (I.P.); (V.V.); (J.G.)
| | - Mila Djisalov
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (M.D.); (L.J.)
| | - Ljiljana Janjušević
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (M.D.); (L.J.)
| | - Jovana Grahovac
- Department of Biotechnology and Pharmaceutical Engineering, Faculty of Technology Novi Sad, University of Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia; (I.P.); (V.V.); (J.G.)
| | - Ivana Gadjanski
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (M.D.); (L.J.)
| |
Collapse
|
28
|
Park JU, Han HJ, Baik JY. Energy metabolism in Chinese hamster ovary (CHO) cells: Productivity and beyond. KOREAN J CHEM ENG 2022. [DOI: 10.1007/s11814-022-1062-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
29
|
MacDonald MA, Barry C, Groves T, Martínez VS, Gray PP, Baker K, Shave E, Mahler S, Munro T, Marcellin E, Nielsen LK. Modelling Apoptosis Resistance in CHO cells with CRISPR-Mediated Knock-outs of Bak1, Bax, and Bok. Biotechnol Bioeng 2022; 119:1380-1391. [PMID: 35180317 PMCID: PMC9310834 DOI: 10.1002/bit.28062] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 12/02/2022]
Abstract
Chinese hamster ovary (CHO) cells are the primary platform for the production of biopharmaceuticals. To increase yields, many CHO cell lines have been genetically engineered to resist cell death. However, the kinetics that governs cell fate in bioreactors are confounded by many variables associated with batch processes. Here, we used CRISPR‐Cas9 to create combinatorial knockouts of the three known BCL‐2 family effector proteins: Bak1, Bax, and Bok. To assess the response to apoptotic stimuli, cell lines were cultured in the presence of four cytotoxic compounds with different mechanisms of action. A population‐based model was developed to describe the behavior of the resulting viable cell dynamics as a function of genotype and treatment. Our results validated the synergistic antiapoptotic nature of Bak1 and Bax, while the deletion of Bok had no significant impact. Importantly, the uniform application of apoptotic stresses permitted direct observation and quantification of a delay in the onset of cell death through Bayesian inference of meaningful model parameters. In addition to the classical death rate, a delay function was found to be essential in the accurate modeling of the cell death response. These findings represent an important bridge between cell line engineering strategies and biological modeling in a bioprocess context.
Collapse
Affiliation(s)
- Michael A MacDonald
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Craig Barry
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Teddy Groves
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs, Lyngby, Denmark
| | - Verónica S Martínez
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Peter P Gray
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Kym Baker
- Patheon by Thermo Fisher Scientific, Woolloongabba, Queensland, 4102, Australia
| | - Evan Shave
- Patheon by Thermo Fisher Scientific, Woolloongabba, Queensland, 4102, Australia
| | - Stephen Mahler
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Trent Munro
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Esteban Marcellin
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia.,Metabolomics Australia, The University of Queensland, Brisbane, Queensland, Australia
| | - Lars K Nielsen
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia.,The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs, Lyngby, Denmark
| |
Collapse
|
30
|
Walsh I, Myint M, Nguyen-Khuong T, Ho YS, Ng SK, Lakshmanan M. Harnessing the potential of machine learning for advancing "Quality by Design" in biomanufacturing. MAbs 2022; 14:2013593. [PMID: 35000555 PMCID: PMC8744891 DOI: 10.1080/19420862.2021.2013593] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ensuring consistent high yields and product quality are key challenges in biomanufacturing. Even minor deviations in critical process parameters (CPPs) such as media and feed compositions can significantly affect product critical quality attributes (CQAs). To identify CPPs and their interdependencies with product yield and CQAs, design of experiments, and multivariate statistical approaches are typically used in industry. Although these models can predict the effect of CPPs on product yield, there is room to improve CQA prediction performance by capturing the complex relationships in high-dimensional data. In this regard, machine learning (ML) approaches offer immense potential in handling non-linear datasets and thus are able to identify new CPPs that could effectively predict the CQAs. ML techniques can also be synergized with mechanistic models as a ‘hybrid ML’ or ‘white box ML’ to identify how CPPs affect the product yield and quality mechanistically, thus enabling rational design and control of the bioprocess. In this review, we describe the role of statistical modeling in Quality by Design (QbD) for biomanufacturing, and provide a generic outline on how relevant ML can be used to meaningfully analyze bioprocessing datasets. We then offer our perspectives on how relevant use of ML can accelerate the implementation of systematic QbD within the biopharma 4.0 paradigm.
Collapse
Affiliation(s)
- Ian Walsh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Matthew Myint
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Terry Nguyen-Khuong
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ying Swan Ho
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Say Kong Ng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore.,Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| |
Collapse
|
31
|
Tihanyi B, Nyitray L. Recent advances in CHO cell line development for recombinant protein production. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 38:25-34. [PMID: 34895638 DOI: 10.1016/j.ddtec.2021.02.003] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/02/2021] [Accepted: 02/23/2021] [Indexed: 12/20/2022]
Abstract
Recombinant proteins used in biomedical research, diagnostics and different therapies are mostly produced in Chinese hamster ovary cells in the pharmaceutical industry. These biotherapeutics, monoclonal antibodies in particular, have shown remarkable market growth in the past few decades. The increasing demand for high amounts of biologics requires continuous optimization and improvement of production technologies. Research aims at discovering better means and methods for reaching higher volumetric capacity, while maintaining stable product quality. An increasing number of complex novel protein therapeutics, such as viral antigens, vaccines, bi- and tri-specific monoclonal antibodies, are currently entering industrial production pipelines. These biomolecules are, in many cases, difficult to express and require tailored product-specific solutions to improve their transient or stable production. All these requirements boost the development of more efficient expression optimization systems and high-throughput screening platforms to facilitate the design of product-specific cell line engineering and production strategies. In this minireview, we provide an overview on recent advances in CHO cell line development, targeted genome manipulation techniques, selection systems and screening methods currently used in recombinant protein production.
Collapse
Affiliation(s)
- Borbála Tihanyi
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter stny 1/C, 1117 Budapest, Hungary
| | - László Nyitray
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter stny 1/C, 1117 Budapest, Hungary.
| |
Collapse
|
32
|
Antonakoudis A, Strain B, Barbosa R, Jimenez del Val I, Kontoravdi C. Synergising stoichiometric modelling with artificial neural networks to predict antibody glycosylation patterns in Chinese hamster ovary cells. Comput Chem Eng 2021. [DOI: 10.1016/j.compchemeng.2021.107471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
33
|
Park SY, Park CH, Choi DH, Hong JK, Lee DY. Bioprocess digital twins of mammalian cell culture for advanced biomanufacturing. Curr Opin Chem Eng 2021. [DOI: 10.1016/j.coche.2021.100702] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Savizi ISP, Motamedian E, E Lewis N, Jimenez Del Val I, Shojaosadati SA. An integrated modular framework for modeling the effect of ammonium on the sialylation process of monoclonal antibodies produced by CHO cells. Biotechnol J 2021; 16:e2100019. [PMID: 34021707 DOI: 10.1002/biot.202100019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Monoclonal antibodies (mABs) have emerged as one of the most important therapeutic recombinant proteins in the pharmaceutical industry. Their immunogenicity and therapeutic efficacy are influenced by post-translational modifications, specifically the glycosylation process. Bioprocess conditions can influence the intracellular process of glycosylation. Among all the process conditions that have been recognized to affect the mAB glycoforms, the detailed mechanism underlying how ammonium could perturb glycosylation remains to be fully understood. It was shown that ammonium induces heterogeneity in protein glycosylation by altering the sialic acid content of glycoproteins. Hence, understanding this mechanism would aid pharmaceutical manufacturers to ensure consistent protein glycosylation. METHODS Three different mechanisms have been proposed to explain how ammonium influences the sialylation process. In the first, the inhibition of CMP-sialic acid transporter, which transports CMP-sialic acid (sialylation substrate) into the Golgi, by an increase in UDP-GlcNAc content that is brought about by the augmented incorporation of ammonium into glucosamine formation. In the second, ammonia diffuses into the Golgi and raises its pH, thereby decreasing the sialyltransferase enzyme activity. In the third, the reduction of sialyltransferase enzyme expression level in the presence of ammonium. We employed these mechanisms in a novel integrated modular platform to link dynamic alteration in mAB sialylation process with extracellular ammonium concentration to elucidate how ammonium alters the sialic acid content of glycoproteins. RESULTS Our results show that the sialylation reaction rate is insensitive to the first mechanism. At low ammonium concentration, the second mechanism is the controlling mechanism in mAB sialylation and by increasing the ammonium level (< 8 mM) the third mechanism becomes the controlling mechanism. At higher ammonium concentrations (> 8 mM) the second mechanism becomes predominant again. CONCLUSION The presented model in this study provides a connection between extracellular ammonium and the monoclonal antibody sialylation process. This computational tool could help scientists to develop and formulate cell culture media. The model illustrated here can assist the researchers to select culture media that ensure consistent mAB sialylation.
Collapse
Affiliation(s)
- Iman Shahidi Pour Savizi
- Faculty of Chemical Engineering, Biotechnology Department, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Motamedian
- Faculty of Chemical Engineering, Biotechnology Department, Tarbiat Modares University, Tehran, Iran
| | - Nathan E Lewis
- Department of Bioengineering, University of California, La Jolla, California, USA.,School of Medicine, Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, California, USA.,Department of Pediatrics, School of Medicine, University of California, La Jolla, California, USA
| | | | - Seyed Abbas Shojaosadati
- Faculty of Chemical Engineering, Biotechnology Department, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
35
|
Nguyen TN, Sha S, Hong MS, Maloney AJ, Barone PW, Neufeld C, Wolfrum J, Springs SL, Sinskey AJ, Braatz RD. Mechanistic model for production of recombinant adeno-associated virus via triple transfection of HEK293 cells. Mol Ther Methods Clin Dev 2021; 21:642-655. [PMID: 34095346 PMCID: PMC8143981 DOI: 10.1016/j.omtm.2021.04.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/08/2021] [Indexed: 02/08/2023]
Abstract
Manufacturing of recombinant adeno-associated virus (rAAV) viral vectors remains challenging, with low yields and low full:empty capsid ratios in the harvest. To elucidate the dynamics of recombinant viral production, we develop a mechanistic model for the synthesis of rAAV viral vectors by triple plasmid transfection based on the underlying biological processes derived from wild-type AAV. The model covers major steps starting from exogenous DNA delivery to the reaction cascade that forms viral proteins and DNA, which subsequently result in filled capsids, and the complex functions of the Rep protein as a regulator of the packaging plasmid gene expression and a catalyst for viral DNA packaging. We estimate kinetic parameters using dynamic data from literature and in-house triple transient transfection experiments. Model predictions of productivity changes as a result of the varied input plasmid ratio are benchmarked against transfection data from the literature. Sensitivity analysis suggests that (1) the poorly coordinated timeline of capsid synthesis and viral DNA replication results in a low ratio of full virions in harvest, and (2) repressive function of the Rep protein could be impeding capsid production at a later phase. The analyses from the mathematical model provide testable hypotheses for evaluation and reveal potential process bottlenecks that can be investigated.
Collapse
Affiliation(s)
- Tam N.T. Nguyen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sha Sha
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Moo Sun Hong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew J. Maloney
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Paul W. Barone
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caleb Neufeld
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacqueline Wolfrum
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stacy L. Springs
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anthony J. Sinskey
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Richard D. Braatz
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
36
|
Tsopanoglou A, Jiménez del Val I. Moving towards an era of hybrid modelling: advantages and challenges of coupling mechanistic and data-driven models for upstream pharmaceutical bioprocesses. Curr Opin Chem Eng 2021. [DOI: 10.1016/j.coche.2021.100691] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
37
|
Štor J, Ruckerbauer DE, Széliová D, Zanghellini J, Borth N. Towards rational glyco-engineering in CHO: from data to predictive models. Curr Opin Biotechnol 2021; 71:9-17. [PMID: 34048995 DOI: 10.1016/j.copbio.2021.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 12/22/2022]
Abstract
Metabolic modelling strives to develop modelling approaches that are robust and highly predictive. To achieve this, various modelling designs, including hybrid models, and parameter estimation methods that define the type and number of parameters used in the model, are adapted. Accurate input data play an important role so that the selection of experimental methods that provide input data of the required precision with low measurement errors is crucial. For the biopharmaceutically relevant protein glycosylation, the most prominent available models are kinetic models which are able to capture the dynamic nature of protein N-glycosylation. In this review we focus on how to choose the most suitable model for a specific research question, as well as on parameters and considerations to take into account before planning relevant experiments.
Collapse
Affiliation(s)
- Jerneja Štor
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, A-1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria
| | - David E Ruckerbauer
- acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria; Department of Analytical Chemistry, University of Vienna, A-1090 Vienna, Austria
| | - Diana Széliová
- acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria; Department of Analytical Chemistry, University of Vienna, A-1090 Vienna, Austria
| | - Jürgen Zanghellini
- acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria; Department of Analytical Chemistry, University of Vienna, A-1090 Vienna, Austria.
| | - Nicole Borth
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, A-1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, A-8010 Graz, Austria.
| |
Collapse
|
38
|
Ben Yahia B, Malphettes L, Heinzle E. Predictive macroscopic modeling of cell growth, metabolism and monoclonal antibody production: Case study of a CHO fed-batch production. Metab Eng 2021; 66:204-216. [PMID: 33887460 DOI: 10.1016/j.ymben.2021.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/08/2021] [Accepted: 04/07/2021] [Indexed: 12/31/2022]
Abstract
We describe a systematic approach to establish predictive models of CHO cell growth, cell metabolism and monoclonal antibody (mAb) formation during biopharmaceutical production. The prediction is based on a combination of an empirical metabolic model connecting extracellular metabolic fluxes with cellular growth and product formation with mixed Monod-inhibition type kinetics that we generalized to every possible external metabolite. We describe the maximum specific growth rate as a function of the integral viable cell density (IVCD). Moreover, we also take into account the accumulation of metabolites in intracellular pools that can influence cell growth. This is possible even without identification and quantification of these metabolites as illustrated with fed-batch cultures of Chinese Hamster Ovary (CHO) cells producing a mAb. The impact of cysteine and tryptophan on cell growth and cell productivity was assessed, and the resulting macroscopic model was successfully used to predict the impact of new, untested feeding strategies on cell growth and mAb production. This model combining piecewise linear relationships between metabolic rates, growth rate and production rate together with Monod-inhibition type models for cell growth did well in predicting cell culture performance in fed-batch cultures even outside the range of experimental data used for establishing the model. It could therefore also successfully be applied for in silico prediction of optimal operating conditions.
Collapse
Affiliation(s)
- Bassem Ben Yahia
- Biochemical Engineering Institute, Saarland University, Campus A1.5, D-66123 Saarbrücken (Germany); Upstream Process Sciences Biotech Sciences, UCB Pharma S.A., Avenue de l'Industrie, Braine l'Alleud B-1420 (Belgium)
| | - Laetitia Malphettes
- Upstream Process Sciences Biotech Sciences, UCB Pharma S.A., Avenue de l'Industrie, Braine l'Alleud B-1420 (Belgium)
| | - Elmar Heinzle
- Biochemical Engineering Institute, Saarland University, Campus A1.5, D-66123 Saarbrücken (Germany).
| |
Collapse
|
39
|
Manstein F, Ullmann K, Kropp C, Halloin C, Triebert W, Franke A, Farr CM, Sahabian A, Haase A, Breitkreuz Y, Peitz M, Brüstle O, Kalies S, Martin U, Olmer R, Zweigerdt R. High density bioprocessing of human pluripotent stem cells by metabolic control and in silico modeling. Stem Cells Transl Med 2021; 10:1063-1080. [PMID: 33660952 PMCID: PMC8235132 DOI: 10.1002/sctm.20-0453] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 12/13/2022] Open
Abstract
To harness the full potential of human pluripotent stem cells (hPSCs) we combined instrumented stirred tank bioreactor (STBR) technology with the power of in silico process modeling to overcome substantial, hPSC‐specific hurdles toward their mass production. Perfused suspension culture (3D) of matrix‐free hPSC aggregates in STBRs was applied to identify and control process‐limiting parameters including pH, dissolved oxygen, glucose and lactate levels, and the obviation of osmolality peaks provoked by high density culture. Media supplements promoted single cell‐based process inoculation and hydrodynamic aggregate size control. Wet lab‐derived process characteristics enabled predictive in silico modeling as a new rational for hPSC cultivation. Consequently, hPSC line‐independent maintenance of exponential cell proliferation was achieved. The strategy yielded 70‐fold cell expansion in 7 days achieving an unmatched density of 35 × 106 cells/mL equivalent to 5.25 billion hPSC in 150 mL scale while pluripotency, differentiation potential, and karyotype stability was maintained. In parallel, media requirements were reduced by 75% demonstrating the outstanding increase in efficiency. Minimal input to our in silico model accurately predicts all main process parameters; combined with calculation‐controlled hPSC aggregation kinetics, linear process upscaling is also enabled and demonstrated for up to 500 mL scale in an independent bioreactor system. Thus, by merging applied stem cell research with recent knowhow from industrial cell fermentation, a new level of hPSC bioprocessing is revealed fueling their automated production for industrial and therapeutic applications.
Collapse
Affiliation(s)
- Felix Manstein
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Kevin Ullmann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Christina Kropp
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Clara-Milena Farr
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Anais Sahabian
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Alexandra Haase
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Yannik Breitkreuz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany.,Cell Programming Core Facility, University of Bonn Medical Faculty, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Stefan Kalies
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| |
Collapse
|
40
|
Modelling Cell Metabolism: A Review on Constraint-Based Steady-State and Kinetic Approaches. Processes (Basel) 2021. [DOI: 10.3390/pr9020322] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Studying cell metabolism serves a plethora of objectives such as the enhancement of bioprocess performance, and advancement in the understanding of cell biology, of drug target discovery, and in metabolic therapy. Remarkable successes in these fields emerged from heuristics approaches, for instance, with the introduction of effective strategies for genetic modifications, drug developments and optimization of bioprocess management. However, heuristics approaches have showed significant shortcomings, such as to describe regulation of metabolic pathways and to extrapolate experimental conditions. In the specific case of bioprocess management, such shortcomings limit their capacity to increase product quality, while maintaining desirable productivity and reproducibility levels. For instance, since heuristics approaches are not capable of prediction of the cellular functions under varying experimental conditions, they may lead to sub-optimal processes. Also, such approaches used for bioprocess control often fail in regulating a process under unexpected variations of external conditions. Therefore, methodologies inspired by the systematic mathematical formulation of cell metabolism have been used to address such drawbacks and achieve robust reproducible results. Mathematical modelling approaches are effective for both the characterization of the cell physiology, and the estimation of metabolic pathways utilization, thus allowing to characterize a cell population metabolic behavior. In this article, we present a review on methodology used and promising mathematical modelling approaches, focusing primarily to investigate metabolic events and regulation. Proceeding from a topological representation of the metabolic networks, we first present the metabolic modelling approaches that investigate cell metabolism at steady state, complying to the constraints imposed by mass conservation law and thermodynamics of reactions reversibility. Constraint-based models (CBMs) are reviewed highlighting the set of assumed optimality functions for reaction pathways. We explore models simulating cell growth dynamics, by expanding flux balance models developed at steady state. Then, discussing a change of metabolic modelling paradigm, we describe dynamic kinetic models that are based on the mathematical representation of the mechanistic description of nonlinear enzyme activities. In such approaches metabolic pathway regulations are considered explicitly as a function of the activity of other components of metabolic networks and possibly far from the metabolic steady state. We have also assessed the significance of metabolic model parameterization in kinetic models, summarizing a standard parameter estimation procedure frequently employed in kinetic metabolic modelling literature. Finally, some optimization practices used for the parameter estimation are reviewed.
Collapse
|
41
|
Bezjak L, Erklavec Zajec V, Baebler Š, Stare T, Gruden K, Pohar A, Novak U, Likozar B. Incorporating RNA-Seq transcriptomics into glycosylation-integrating metabolic network modelling kinetics: Multiomic Chinese hamster ovary (CHO) cell bioreactors. Biotechnol Bioeng 2021; 118:1476-1490. [PMID: 33399226 DOI: 10.1002/bit.27660] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/10/2020] [Accepted: 11/16/2020] [Indexed: 12/23/2022]
Abstract
In this work, the kinetic model based on the previously developed metabolic and glycan reaction networks of the ovarian cells of the Chinese hamster ovary (CHO) cell line was improved by the inclusion of transcriptomic data that took into account the values of the RPKM gene (Reads per Kilobase of Exon per Million Reads Mapped). The transcriptomic (RNASeq) data were obtained together with metabolic and glycan data from the literature, and the concentrations with RPKM values were collected at several points in time from two fed-batch processes. First, the fluxes were determined by regression analysis of the metabolic data, then these fluxes were corrected by using the fold change in gene expression as a measure of enzyme concentrations. Next, the corrected fluxes in the kinetic model were used to calculate the concentration profiles of the metabolites, and literature data were used to evaluate the predicted results of the model. Compared to other studies where the concentration profiles of CHO cell metabolites were described using a kinetic model without consideration of RNA-Seq data to correct the fluxes, this model is unique. The additional integration of transcriptomic data led to better predictions of metabolic concentrations in the fed-batch process, which is a significant improvement of the modelling technique used.
Collapse
Affiliation(s)
- Lara Bezjak
- Department of Catalysis and Chemical Reaction Engineering, National Institute of Chemistry, Ljubljana, Slovenia
| | - Vivian Erklavec Zajec
- Department of Catalysis and Chemical Reaction Engineering, National Institute of Chemistry, Ljubljana, Slovenia
| | - Špela Baebler
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Tjaša Stare
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Kristina Gruden
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Andrej Pohar
- Department of Catalysis and Chemical Reaction Engineering, National Institute of Chemistry, Ljubljana, Slovenia
| | - Uroš Novak
- Department of Catalysis and Chemical Reaction Engineering, National Institute of Chemistry, Ljubljana, Slovenia
| | - Blaž Likozar
- Department of Catalysis and Chemical Reaction Engineering, National Institute of Chemistry, Ljubljana, Slovenia
| |
Collapse
|
42
|
Karimian E, Motamedian E. ACBM: An Integrated Agent and Constraint Based Modeling Framework for Simulation of Microbial Communities. Sci Rep 2020; 10:8695. [PMID: 32457521 PMCID: PMC7250870 DOI: 10.1038/s41598-020-65659-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 05/05/2020] [Indexed: 12/25/2022] Open
Abstract
The development of new methods capable of more realistic modeling of microbial communities necessitates that their results be quantitatively comparable with experimental findings. In this research, a new integrated agent and constraint based modeling framework abbreviated ACBM has been proposed that integrates agent-based and constraint-based modeling approaches. ACBM models the cell population in three-dimensional space to predict spatial and temporal dynamics and metabolic interactions. When used to simulate the batch growth of C. beijerinckii and two-species communities of F. prausnitzii and B. adolescent., ACBM improved on predictions made by two previous models. Furthermore, when transcriptomic data were integrated with a metabolic model of E. coli to consider intracellular constraints in the metabolism, ACBM accurately predicted growth rate, half-rate constant, and concentration of biomass, glucose, and acidic products over time. The results also show that the framework was able to predict the metabolism changes in the early stationary compared to the log phase. Finally, ACBM was implemented to estimate starved cells under heterogeneous feeding and it was concluded that a percentage of cells are always subject to starvation in a bioreactor with high volume.
Collapse
Affiliation(s)
- Emadoddin Karimian
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, P.O. Box, 14115-143, Tehran, Iran
| | - Ehsan Motamedian
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, P.O. Box, 14115-143, Tehran, Iran.
| |
Collapse
|
43
|
Yeo HC, Hong J, Lakshmanan M, Lee DY. Enzyme capacity-based genome scale modelling of CHO cells. Metab Eng 2020; 60:138-147. [PMID: 32330653 DOI: 10.1016/j.ymben.2020.04.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/21/2020] [Accepted: 04/14/2020] [Indexed: 10/24/2022]
Abstract
Chinese hamster ovary (CHO) cells are most prevalently used for producing recombinant therapeutics in biomanufacturing. Recently, more rational and systems approaches have been increasingly exploited to identify key metabolic bottlenecks and engineering targets for cell line engineering and process development based on the CHO genome-scale metabolic model which mechanistically characterizes cell culture behaviours. However, it is still challenging to quantify plausible intracellular fluxes and discern metabolic pathway usages considering various clonal traits and bioprocessing conditions. Thus, we newly incorporated enzyme kinetic information into the updated CHO genome-scale model (iCHO2291) and added enzyme capacity constraints within the flux balance analysis framework (ecFBA) to significantly reduce the flux variability in biologically meaningful manner, as such improving the accuracy of intracellular flux prediction. Interestingly, ecFBA could capture the overflow metabolism under the glucose excess condition where the usage of oxidative phosphorylation is limited by the enzyme capacity. In addition, its applicability was successfully demonstrated via a case study where the clone- and media-specific lactate metabolism was deciphered, suggesting that the lactate-pyruvate cycling could be beneficial for CHO cells to efficiently utilize the mitochondrial redox capacity. In summary, iCHO2296 with ecFBA can be used to confidently elucidate cell cultures and effectively identify key engineering targets, thus guiding bioprocess optimization and cell engineering efforts as a part of digital twin model for advanced biomanufacturing in future.
Collapse
Affiliation(s)
- Hock Chuan Yeo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, 138668, Singapore; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore
| | - Jongkwang Hong
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, 138668, Singapore
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, 138668, Singapore.
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, 138668, Singapore; School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
| |
Collapse
|
44
|
A PSE perspective for the efficient production of monoclonal antibodies: integration of process, cell, and product design aspects. Curr Opin Chem Eng 2020. [DOI: 10.1016/j.coche.2020.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
45
|
Ramos JRC, Rath AG, Genzel Y, Sandig V, Reichl U. A dynamic model linking cell growth to intracellular metabolism and extracellular by-product accumulation. Biotechnol Bioeng 2020; 117:1533-1553. [PMID: 32022250 DOI: 10.1002/bit.27288] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 12/12/2019] [Accepted: 01/26/2020] [Indexed: 12/16/2022]
Abstract
Mathematical modeling of animal cell growth and metabolism is essential for the understanding and improvement of the production of biopharmaceuticals. Models can explain the dynamic behavior of cell growth and product formation, support the identification of the most relevant parameters for process design, and significantly reduce the number of experiments to be performed for process optimization. Few dynamic models have been established that describe both extracellular and intracellular dynamics of growth and metabolism of animal cells. In this study, a model was developed, which comprises a set of 33 ordinary differential equations to describe batch cultivations of suspension AGE1.HN.AAT cells considered for the production of α1-antitrypsin. This model combines a segregated cell growth model with a structured model of intracellular metabolism. Overall, it considers the viable cell concentration, mean cell diameter, viable cell volume, concentration of extracellular substrates, and intracellular concentrations of key metabolites from the central carbon metabolism. Furthermore, the release of metabolic by-products such as lactate and ammonium was estimated directly from the intracellular reactions. Based on the same set of parameters, this model simulates well the dynamics of four independent batch cultivations. Analysis of the simulated intracellular rates revealed at least two distinct cellular physiological states. The first physiological state was characterized by a high glycolytic rate and high lactate production. Whereas the second state was characterized by efficient adenosine triphosphate production, a low glycolytic rate, and reactions of the TCA cycle running in the reverse direction from α-ketoglutarate to citrate. Finally, we show possible applications of the model for cell line engineering and media optimization with two case studies.
Collapse
Affiliation(s)
- João R C Ramos
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Alexander G Rath
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- Bioprocess Engineering, AMINO GmbH, Frellstedt, Germany
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Volker Sandig
- Bioprocess Engineering, ProBioGen AG, Berlin, Germany
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- Bioprocess Engineering, Otto von Guericke University Magdeburg, Chair of Bioprocess Engineering, Magdeburg, Germany
| |
Collapse
|
46
|
Tang P, Xu J, Louey A, Tan Z, Yongky A, Liang S, Li ZJ, Weng Y, Liu S. Kinetic modeling of Chinese hamster ovary cell culture: factors and principles. Crit Rev Biotechnol 2020; 40:265-281. [DOI: 10.1080/07388551.2019.1711015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Peifeng Tang
- Department of Paper and Bioprocess Engineering, SUNY-ESF, Syracuse, NY, USA
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| | - Jianlin Xu
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| | - Alastair Louey
- Elpiscience Biopharma, Cayman Islands George Town, Grand Cayman, UK
| | - Zhijun Tan
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| | - Andrew Yongky
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| | - Shaoyan Liang
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT, USA
| | - Zheng Jian Li
- Global Product Development and Supply, Bristol-Myers Squibb Company, Devens, MA, USA
| | - Yongyan Weng
- Department of Civil Engineering, University of Nottingham, Nottingham, UK
| | - Shijie Liu
- Department of Paper and Bioprocess Engineering, SUNY-ESF, Syracuse, NY, USA
| |
Collapse
|
47
|
Villaverde AF, Fröhlich F, Weindl D, Hasenauer J, Banga JR. Benchmarking optimization methods for parameter estimation in large kinetic models. Bioinformatics 2019; 35:830-838. [PMID: 30816929 PMCID: PMC6394396 DOI: 10.1093/bioinformatics/bty736] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/04/2018] [Accepted: 08/21/2018] [Indexed: 11/18/2022] Open
Abstract
Motivation Kinetic models contain unknown parameters that are estimated by optimizing the fit to experimental data. This task can be computationally challenging due to the presence of local optima and ill-conditioning. While a variety of optimization methods have been suggested to surmount these issues, it is difficult to choose the best one for a given problem a priori. A systematic comparison of parameter estimation methods for problems with tens to hundreds of optimization variables is currently missing, and smaller studies provided contradictory findings. Results We use a collection of benchmarks to evaluate the performance of two families of optimization methods: (i) multi-starts of deterministic local searches and (ii) stochastic global optimization metaheuristics; the latter may be combined with deterministic local searches, leading to hybrid methods. A fair comparison is ensured through a collaborative evaluation and a consideration of multiple performance metrics. We discuss possible evaluation criteria to assess the trade-off between computational efficiency and robustness. Our results show that, thanks to recent advances in the calculation of parametric sensitivities, a multi-start of gradient-based local methods is often a successful strategy, but a better performance can be obtained with a hybrid metaheuristic. The best performer combines a global scatter search metaheuristic with an interior point local method, provided with gradients estimated with adjoint-based sensitivities. We provide an implementation of this method to render it available to the scientific community. Availability and implementation The code to reproduce the results is provided as Supplementary Material and is available at Zenodo https://doi.org/10.5281/zenodo.1304034. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Fabian Fröhlich
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Mathematical Modeling of Biological Systems, Center for Mathematics, Technische Universität München, Garching, Germany
| | - Daniel Weindl
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Jan Hasenauer
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Mathematical Modeling of Biological Systems, Center for Mathematics, Technische Universität München, Garching, Germany
| | | |
Collapse
|
48
|
Hong JK, Yeo HC, Lakshmanan M, Han SH, Cha HM, Han M, Lee DY. In silico model-based characterization of metabolic response to harsh sparging stress in fed-batch CHO cell cultures. J Biotechnol 2019; 308:10-20. [PMID: 31756358 DOI: 10.1016/j.jbiotec.2019.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/11/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022]
Abstract
Mammalian cell culture platform has been successfully implemented for industrial biopharmaceutical production through the advancements in early stage process development including cell-line engineering, media design and process optimization. However, late stage developments such as scale-up, scale-down and large-scale cell cultivation still face many industrial challenges to acquire comparable process performance between different culture scales. One of them is the sparging strategy which significantly affects productivity, quality and comparability. Currently, it is mainly relying on the empirical records due to the lack of theoretical framework and scarcity of available literatures to elucidate intracellular metabolic features. Therefore, it is highly required to characterize the underlying mechanism of physiological changes and metabolic states upon the aeration stress. To this end, initially we cultivated antibody producing CHO cells under mild and harsh sparging conditions and observed that sparging stress leads to the decreased cell growth rate, viability and productivity. Subsequent in silicomodel-driven flux analysis suggested that sparging stress rewires amino acid metabolism towards the enriched H2O2 turnover rate by up-regulated fluxes of amino acid oxidases. Interestingly, many of these H2O2-generating reactions were closely connected with the production of NADH, NADPH and GSH which are typical reducing equivalents. Thus, we can hypothesize that increased amino acid uptake caused by sparging stress contributes to restore redox homeostasis against oxidative stress. The current model-driven systematic data analysis allows us to quickly define distinct metabolic feature under stress condition by using basic cell cultivation datasets.
Collapse
Affiliation(s)
- Jong Kwang Hong
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, 138668, Singapore
| | - Hock Chuan Yeo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, 138668, Singapore
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, 138668, Singapore
| | - Sung-Hyuk Han
- Upstream process, GC Pharma R&D center, 107 Ihyun-ro, Giheung-gu, Yongin-si, Gyeonggi-do, 16926, Republic of Korea
| | - Hyun Myoung Cha
- Upstream process, GC Pharma R&D center, 107 Ihyun-ro, Giheung-gu, Yongin-si, Gyeonggi-do, 16926, Republic of Korea
| | - Muri Han
- Upstream process, GC Pharma R&D center, 107 Ihyun-ro, Giheung-gu, Yongin-si, Gyeonggi-do, 16926, Republic of Korea
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, 138668, Singapore; School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
| |
Collapse
|
49
|
Grilo AL, Mantalaris A. A Predictive Mathematical Model of Cell Cycle, Metabolism, and Apoptosis of Monoclonal Antibody‐Producing GS–NS0 Cells. Biotechnol J 2019; 14:e1800573. [DOI: 10.1002/biot.201800573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 06/22/2019] [Indexed: 12/18/2022]
Affiliation(s)
- António L. Grilo
- Biological Systems Engineering Laboratory Department of Chemical Engineering Centre for Process Systems EngineeringImperial College LondonExhibition Road London SW7 2AZ UK
| | - Athanasios Mantalaris
- Biological Systems Engineering Laboratory Department of Chemical Engineering Centre for Process Systems EngineeringImperial College LondonExhibition Road London SW7 2AZ UK
- Wallace H. Coulter Department of Biomedical Engineering Biomedical Systems Engineering LaboratoryGeorgia Institute of Technology950 Atlantic Drive Atlanta GA 30332 USA
| |
Collapse
|
50
|
Heterogeneity Studies of Mammalian Cells for Bioproduction: From Tools to Application. Trends Biotechnol 2019; 37:645-660. [DOI: 10.1016/j.tibtech.2018.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 12/22/2022]
|