Integration Approaches to Model Bioreactor Hydrodynamics and Cellular Kinetics for Advancing Bioprocess Optimisation
Abstract
:1. Introduction
2. Literature Search Methodology
3. CFD-CRK Coupling Approaches
3.1. Eulerian Approach
3.2. Lagrangian Approach
4. Selecting a Suitable Coupling Strategy
5. State of the Art
Reference | System Description | Integration Approach | Validation Method | Year |
---|---|---|---|---|
[88] | Proprietary mammalian cell line, 200 L bioreactor | Euler–Lagrange (EL) simulations with Eulerian reaction coupling. The only reaction to model oxygen and pH profile has been coupled | Modelled Oxygen and pH concentrations verified experimentally | 2024 |
[87] | Escherichia coli and Saccharomyces cerevisiae, 90 m3 bubble column and a stirred tank bioreactor | Euler–Euler simulations with Lagrangian particle tracking approach for cells. Unstructured kinetic model for growth and nutrient uptake rates | Results were completely in silico | 2023 |
[90] | S. cerevisiae, 22 m3 stirred tank reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Unstructured model for nutrient uptake rates | Glucose concentration field data validated against data from literature [85] | 2023 |
[95] | Cell line not mentioned, Perfusion bioreactor volume not mentioned | Euler–Euler simulations with unstructured kinetic model for growth and substrate uptake | Results were completely in silico | 2022 |
[93] | P. chrysogenum, 54 m3 stirred tank reactor | Euler–Lagrange (EL) compartment-based CFD simulations with Lagrangian reaction coupling. Unstructured and structured cellular model for growth rate distribution | The kinetic model was validated against literature data [97], and the CFD-CRK model was validated against mean specific growth rate data from industrial experimental data | 2022 |
[37] | E. coli, 1.5 L stirred-tank bioreactor | Euler–Lagrange (EL) simulations with Eulerian reaction coupling. Unstructured cellular model for the effect of dissolved oxygen on cell growth | Volume average of substrate and product concentrations were validated against experimental data | 2021 |
[94] | Streptococcus thermophilus, 700 L stirred tank reactor | Euler–Euler simulations with unstructured kinetic model for growth and pH change | pH gradient results generated from the CFD-CRK model were experimentally validated using 6 multiple probes at different locations | 2019 |
[36] | Clostridium ljungdahlii, 125 m3 bubble column | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. stoichiometry model for carbon monoxide uptake | Results were completely in silico | 2019 |
[98] | P. chrysogenum, 54 m3 stirred tank reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Structured cellular model for growth rate distribution | The kinetic model was validated against the literature data [97], and the CFD-CRK model was validated against mean specific growth rate data from industrial experimental data | 2018 |
[38] | CHO cell, 3 L bioreactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Unstructured cellular model for the effect of DO on cell growth | Results were completely in silico | 2018 |
[77] | P. chrysogenum, 98 m3 hypothetical reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Unstructured model for nutrient uptake rates | Mean concentration profiles were compared against the literature data | 2017 |
[74] | S. cerevisiae, 22 m3 stirred tank reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Unstructured model for nutrient uptake rates | Glucose concentration field data validated against data from the literature [85] | 2017 |
[75] | Pseudomonas putida, 54 m3 stirred tank reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. An unstructured cellular model involving logistic equation for growth | Results were completely in silico | 2017 |
[96] | HFN 7.1 murine hybridoma cells, 0.01 m3 stirred tank reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Unstructured cellular model for the distribution of cells over cell cycle and effect of pH, dissolved oxygen, gas holdup and energy dissipation rate on cell metabolism | Results were completely in silico | 2017 |
[99] | Carthamus tinctorius L., 5 L–15 L stirred tank reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Unstructured model for death kinetics | CFD results were validated by Particle Image Velocimetry (PIV) measurements, and death kinetics was validated experimentally by viability data. | 2016 |
[100] | S. cerevisiae, 0.24 m3 bubble column reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Unstructured model for nutrient uptake rates | CFD and kinetic models were validated against the literature data [101,102]. No validation of the glucose concentration field. | 2016 |
[70] | Penicillium chrysogenum, 54 m3 stirred tank reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Unstructured model for nutrient uptake rates | Mean substrate concentrations were validated against simulated average substrate concentrations from the Eulerian method. | 2016 |
[72] | E. coli, 22 m3 stirred tank reactor | Euler–Euler simulations with Population balance models for cells. Structured model for growth rate distribution | The volume average of substrate and product concentrations was validated against experimental data and data from the literature [103]. | 2015 |
[71] | Cell line not mentioned, 0.07/70 m3 stirred tank reactor | Euler–Euler simulations with Population balance models for cells. Unstructured model for growth rate distribution | Fluid flow validation from Particle Image Velocimetry (PIV) measurements from the literature [104,105]. No validation for the spatial distribution of specific growth rates. | 2014 |
[73] | Hypothetical aerobic bacteria (like Candida tropicalis), 3-L reactor | Euler–Euler simulations with Population balance models for cells. Unstructured model for nutrient uptake rates | Results generated from the CFD-CRK model were validated against the literature data from [106] | 2013 |
[107] | Aspergillus niger, 5 dm3 stirred-tank bioreactor | Euler–Euler simulations with Eulerian reaction coupling. Unstructured cellular model for growth and product formation | Numerical results are validated against experimental data for a batch time of 60 h | 2013 |
[86] | CHO 320 producing interferon-γ, 1.4 L regulated stirred tank reactor | Empirical Methodology: experimental data fitting to correlate hydrodynamic parameters (mean turbulent energy dissipation rate and Reynolds number) to integral viable cell density | The fluid flow field was validated by Laser Doppler Velocimetry (LDV) measurements. Experimental validation of empirical correlation for hydrodynamic parameters to integral viable cell density | 2010 |
[108] | Fibroblast growth factor-2 (FGF-2) producing endothelial cells, FiberCell® hollow-fibre (bioreactor volume not mentioned) | Euler–Euler simulations with Eulerian reaction coupling. Unstructured cellular model for protein production | Concentration of protein till 600 s at the boundary wall of fibre validated against the experimental data | 2010 |
[83] | Escherichia coli, 0.9 m3 stirred tank reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Structured cellular model applied for sugar uptake | CFD simulations and kinetic model validation with experimental data from literature [109,110]. Glucose concentration field data is qualitatively verified from experimental observations from the literature [85]. | 2006 |
[78] | Saccharomyces cerevisiae, 68 L and 900 L stirred tank reactor | Euler–Lagrange (EL) simulations with Lagrangian reaction coupling. Structured cellular model applied for glycolysis pathway | Lagrange strategy validation by mixing time experiment with tracer substance. No validation for oscillating yeast cells in spacetime | 2004 |
[85] | Saccharomyces cerevisiae, 30 m3 stirred tank reactor | Euler–Euler simulations with Eulerian reaction coupling. Unstructured cellular model for growth and product formation | Hydrodynamic parameters were validated against experimental data from thermal anemometry. Nutrient concentrations were validated against data collected from 3 sensors at different locations | 1996 |
6. Conclusions and Future Prospects
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Nomenclature
Abbreviations | |
USD | United States Dollar |
CFD | Computational fluid dynamics |
CRK | Cell reaction kinetic |
DO | Dissolved oxygen |
EE | Euler-Euler |
EL | Euler-Lagrange |
LDV | Laser Doppler Velocimetry |
PBMs | Population balance models |
PIV | Particle Image Velocimetry |
rCFD | Recurrence-CFD |
TVD | Total Variation Diminishing |
Symbols | |
µ | Viscosity of the liquid (cP) |
Di | Impeller diameter (m) |
dp | Particle diameter (m) |
kLa | Volumetric mass transfer coefficient (1/h) |
L | Distance between particles/characteristic system length (m) |
N | Agitation rate |
P/V | Power density (W/m3) |
St | Stokes number |
v | Characteristic velocity (m/s) |
λk | Kolmogorov length (μm) |
ρ | Density of liquid (kg/m3) |
ρ′ | Density of dispersed or particulate phase (kg/m3) |
τ | Shear stress (N/m2) |
References
- Mordor Intelligence. Biopharmaceutical Industry Size & Share Analysis—Growth Trends & Forecasts (2024–2029). Available online: https://www.mordorintelligence.com/industry-reports/global-biopharmaceuticals-market-industry (accessed on 13 February 2024).
- Evaluate Pharma. World Preview 2021, Outlook to 2026. Available online: https://info.evaluate.com/rs/607-YGS-364/images/WorldPreviewReport_Final_2021.pdf (accessed on 27 March 2024).
- Wang, G.; Tang, W.; Xia, J.; Chu, J.; Noorman, H.; Gulik, W.M. Integration of microbial kinetics and fluid dynamics toward model-driven scale-up of industrial bioprocesses. Eng. Life Sci. 2015, 15, 20–29. [Google Scholar] [CrossRef]
- Lara, A.R.; Galindo, E.; Ramírez, O.T.; Palomares, L.A. Living with heterogeneities in bioreactors: Understanding the effects of environmental gradients on cells. Mol. Biotechnol. 2006, 34, 355–381. [Google Scholar] [CrossRef] [PubMed]
- Enfors, S.-O.; Jahic, M.; Rozkov, A.; Xu, B.; Hecker, M.; Jürgen, B.; Krüger, E.; Schweder, T.; Hamer, G.; O’Beirne, D.; et al. Physiological responses to mixing in large scale bioreactors. J. Biotechnol. 2001, 85, 175–185. [Google Scholar] [CrossRef] [PubMed]
- Hewitt, C.J.; Caron, G.N.-V.; Nienow, A.W.; McFarlane, C.M. The use of multi-parameter flow cytometry to compare the physiological response of Escherichia coli W3110 to glucose limitation during batch, fed-batch and continuous culture cultivations. J. Biotechnol. 1999, 75, 251–264. [Google Scholar] [CrossRef] [PubMed]
- Kuschel, M.; Takors, R. Simulated oxygen and glucose gradients as a prerequisite for predicting industrial scale performance a priori. Biotechnol. Bioeng. 2020, 117, 2760–2770. [Google Scholar] [CrossRef] [PubMed]
- Anane, E.; Knudsen, I.M.; Wilson, G.C. Scale-down cultivation in mammalian cell bioreactors—The effect of bioreactor mixing time on the response of CHO cells to dissolved oxygen gradients. Biochem. Eng. J. 2021, 166, 107870. [Google Scholar] [CrossRef]
- Restelli, V.; Wang, M.D.; Huzel, N.; Ethier, M.; Perreault, H.; Butler, M. The effect of dissolved oxygen on the production and the glycosylation profile of recombinant human erythropoietin produced from CHO cells. Biotechnol. Bioeng. 2006, 94, 481–494. [Google Scholar] [CrossRef] [PubMed]
- Ivarsson, M.; Villiger, T.K.; Morbidelli, M.; Soos, M. Evaluating the impact of cell culture process parameters on monoclonal antibody N-glycosylation. J. Biotechnol. 2014, 188, 88–96. [Google Scholar] [CrossRef] [PubMed]
- Hansen, G.; Johansen, C.L.; Marten, G.; Wilmes, J.; Jespersen, L.; Arneborg, N. Influence of extracellular pH on growth, viability, cell size, acidification activity, and intracellular pH of Lactococcus lactis in batch fermentations. Appl. Microbiol. Biotechnol. 2016, 100, 5965–5976. [Google Scholar] [CrossRef]
- Michl, J.; Park, K.C.; Swietach, P. Evidence-based guidelines for controlling pH in mammalian live-cell culture systems. Commun. Biol. 2019, 2, 1–12. [Google Scholar] [CrossRef]
- Neubauer, P.; Junne, S. Scale-down simulators for metabolic analysis of large-scale bioprocesses. Curr. Opin. Biotechnol. 2010, 21, 114–121. [Google Scholar] [CrossRef] [PubMed]
- Delvigne, F.; Goffin, P. Microbial heterogeneity affects bioprocess robustness: Dynamic single-cell analysis contributes to understanding of microbial populations. Biotechnol. J. 2014, 9, 61–72. [Google Scholar] [CrossRef] [PubMed]
- Borys, M.C.; Dalal, N.G.; Abu-Absi, N.R.; Khattak, S.F.; Jing, Y.; Xing, Z.; Li, Z.J. Effects of culture conditions on N-glycolylneuraminic acid (Neu5Gc) content of a recombinant fusion protein produced in CHO cells. Biotechnol. Bioeng. 2010, 105, 1048–1057. [Google Scholar] [CrossRef] [PubMed]
- Bonarius, H.P.J.; de Gooijer, C.D.; Tramper, J.; Schmid, G. Determination of the respiration quotient in mammalian cell culture in bicarbonate buffered media. Biotechnol. Bioeng. 1995, 45, 524–535. [Google Scholar] [CrossRef]
- Olsson, L.; Rugbjerg, P.; Pianale, L.T.; Trivellin, C. Robustness: Linking strain design to viable bioprocesses. Trends Biotechnol. 2022, 40, 918–931. [Google Scholar] [CrossRef] [PubMed]
- Gaugler, L.; Mast, Y.; Fitschen, J.; Hofmann, S.; Schlüter, M.; Takors, R. Scaling-down biopharmaceutical production processes via a single multi-compartment bioreactor (SMCB). Eng. Life Sci. 2023, 23, e2100161. [Google Scholar] [CrossRef] [PubMed]
- Haringa, C.; Mudde, R.F.; Noorman, H.J. From industrial fermentor to CFD-guided downscaling: What have we learned? Biochem. Eng. J. 2018, 140, 57–71. [Google Scholar] [CrossRef]
- Nadal-Rey, G.; McClure, D.D.; Kavanagh, J.M.; Cornelissen, S.; Fletcher, D.F.; Gernaey, K.V. Understanding gradients in industrial bioreactors. Biotechnol. Adv. 2020, 46, 107660. [Google Scholar] [CrossRef]
- Pramparo, L.; Pruvost, J.; Stüber, F.; Font, J.; Fortuny, A.; Fabregat, A.; Legentilhomme, P.; Legrand, J.; Bengoa, C. Mixing and hydrodynamics investigation using CFD in a square-sectioned torus reactor in batch and continuous regimes. Chem. Eng. J. 2008, 137, 386–395. [Google Scholar] [CrossRef]
- Brannock, M.; Leslie, G.; Wang, Y.; Buetehorn, S. Optimising mixing and nutrient removal in membrane bioreactors: CFD modelling and experimental validation. Desalination 2010, 250, 815–818. [Google Scholar] [CrossRef]
- Singh, V.K. From Fluids to Pharmaceuticals: How Computational Fluid Dynamics (CFD) Transforms Drug Design. Boolean Snapshots Dr. Res. Univ. Coll. Cork 2024, 7, 43–47. [Google Scholar] [CrossRef]
- Ghadge, R.S.; Patwardhan, A.W.; Joshi, J.B. Combined effect of hydrodynamic and interfacial flow parameters on lysozyme deactivation in a stirred tank bioreactor. Biotechnol. Prog. 2006, 22, 660–672. [Google Scholar] [CrossRef]
- Li, X.; Scott, K.; Kelly, W.J.; Huang, Z. Development of a Computational Fluid Dynamics Model for Scaling-up Ambr Bioreactors. Biotechnol. Bioprocess Eng. 2018, 23, 710–725. [Google Scholar] [CrossRef]
- Sucosky, P.; Osorio, D.F.; Brown, J.B.; Neitzel, G.P. Fluid Mechanics of a Spinner-Flask Bioreactor. Biotechnol. Bioeng. 2004, 85, 34–46. [Google Scholar] [CrossRef] [PubMed]
- Williams, K.A.; Saini, S.; Wick, T.M. Computational fluid dynamics modeling of steady-state momentum and mass transport in a bioreactor for cartilage tissue engineering. Biotechnol. Prog. 2002, 18, 951–963. [Google Scholar] [CrossRef] [PubMed]
- Formenti, L.R.; Nørregaard, A.; Bolic, A.; Hernandez, D.Q.; Hagemann, T.; Heins, A.-L.; Larsson, H.; Mears, L.; Mauricio-Iglesias, M.; Krühne, U.; et al. Challenges in industrial fermentation technology research. Biotechnol. J. 2014, 9, 727–738. [Google Scholar] [CrossRef] [PubMed]
- Rotondi, M.; Grace, N.; Betts, J.; Bargh, N.; Costariol, E.; Zoro, B.; Hewitt, C.J.; Nienow, A.W.; Rafiq, Q.A. Design and development of a new ambr250® bioreactor vessel for improved cell and gene therapy applications. Biotechnol. Lett. 2021, 43, 1103–1116. [Google Scholar] [CrossRef]
- Hutmacher, D.W.; Singh, H. Computational fluid dynamics for improved bioreactor design and 3D culture. Trends Biotechnol. 2008, 26, 166–172. [Google Scholar] [CrossRef] [PubMed]
- Scully, J.; Considine, L.B.; Smith, M.T.; McAlea, E.; Jones, N.; O’Connell, E.; Madsen, E.; Power, M.; Mellors, P.; Crowley, J.; et al. Beyond heuristics: CFD-based novel multiparameter scale-up for geometrically disparate bioreactors demonstrated at industrial 2kL–10kL scales. Biotechnol. Bioeng. 2020, 117, 1710–1723. [Google Scholar] [CrossRef]
- Noorman, H.J.; Heijnen, J.J. Biochemical engineering’s grand adventure. Chem. Eng. Sci. 2017, 170, 677–693. [Google Scholar] [CrossRef]
- Fernandes, R.L.; Nierychlo, M.; Lundin, L.; Pedersen, A.E.; Tellez, P.E.; Dutta, A.; Carlquist, M.; Bolic, A.; Schäpper, D.; Brunetti, A.C.; et al. Experimental methods and modeling techniques for description of cell population heterogeneity. Biotechnol. Adv. 2011, 29, 575–599. [Google Scholar] [CrossRef] [PubMed]
- Grima, R.; Schnell, S. Modelling reaction kinetics inside cells. Essays Biochem. 2008, 45, 41. [Google Scholar] [CrossRef] [PubMed]
- Rocha, M.; Neves, J.; Rocha, I.; Ferreira, E.C. Evolutionary algorithms for optimal control in fed-batch fermentation processes. Lect. Notes Comput. Sci. 2004, 3005, 84–93. [Google Scholar] [CrossRef]
- Siebler, F.; Lapin, A.; Hermann, M.; Takors, R. The impact of CO gradients on C. ljungdahlii in a 125 m3 bubble column: Mass transfer, circulation time and lifeline analysis. Chem. Eng. Sci. 2019, 207, 410–423. [Google Scholar] [CrossRef]
- Yeoh, J.W.; Jayaraman, S.S.; Tan, S.G.D.; Jayaraman, P.; Holowko, M.B.; Zhang, J.; Kang, C.W.; Leo, H.L.; Poh, C.L. A model-driven approach towards rational microbial bioprocess optimization. Biotechnol. Bioeng. 2020, 118, 1–35. [Google Scholar] [CrossRef] [PubMed]
- Farzan, P.; Ierapetritou, M.G. A Framework for the Development of Integrated and Computationally Feasible Models of Large-Scale Mammalian Cell Bioreactors. Processes 2018, 6, 82. [Google Scholar] [CrossRef]
- Schmidt, A.; Hengelbrock, A.; Strube, J. Biopharma 4.0 for Biologics Manufacturing Under Pandemic Constraints. In Biopharmaceutical Manufacturing: Progress, Trends and Challenges; Springer: Cham, Switzerland, 2023; pp. 297–317. [Google Scholar] [CrossRef]
- Hansen, S.; Gumprecht, A.; Micheel, L.; Hennemann, H.G.; Enzmann, F.; Blümke, W. Implementation of Perforated Concentric Ring Walls Considerably Improves Gas-Liquid Mass Transfer of Shaken Bioreactors. Front. Bioeng. Biotechnol. 2022, 10, 894295. [Google Scholar] [CrossRef] [PubMed]
- Seidel, S.; Eibl, D.; Wawrzyniak, P.; Jaskulski, M. Influence of Interfacial Force Models and Population Balance Models on the kLa Value in Stirred Bioreactors. Processes 2021, 9, 1185. [Google Scholar] [CrossRef]
- Niño, L.; Gelves, R.; Ali, H.; Solsvik, J.; Jakobsen, H. Applicability of a modified breakage and coalescence model based on the complete turbulence spectrum concept for CFD simulation of gas-liquid mass transfer in a stirred tank reactor. Chem. Eng. Sci. 2020, 211, 115272. [Google Scholar] [CrossRef]
- Maschke, R.W.; Seidel, S.; Bley, T.; Eibl, R.; Eibl, D. Determination of culture design spaces in shaken disposable cultivation systems for CHO suspension cell cultures. Biochem. Eng. J. 2022, 177, 108224. [Google Scholar] [CrossRef]
- Thomas, J.A.; Liu, X.; DeVincentis, B.; Hua, H.; Yao, G.; Borys, M.C.; Aron, K.; Pendse, G. A mechanistic approach for predicting mass transfer in bioreactors. Chem. Eng. Sci. 2021, 237, 116538. [Google Scholar] [CrossRef]
- Svay, K.; Urrea, C.; Shamlou, P.A.; Zhang, H. Computational fluid dynamics analysis of mixing and gas–liquid mass transfer in wave bag bioreactor. Biotechnol. Prog. 2020, 36, e3049. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.; Jurtz, N.; Walle, A.; Kraume, M. Evaluation and application of efficient CFD-based methods for the multi-objective optimization of stirred tanks. Chem. Eng. Sci. 2022, 263, 118109. [Google Scholar] [CrossRef]
- Madhania, S.; Fathonah, N.N.; Kusdianto; Nurtono, T.; Winardi, S. Turbulence Modeling in Side-Entry Stirred Tank Mixing Time Determination. MATEC Web Conf. 2021, 333, 02003. [Google Scholar] [CrossRef]
- Wutz, J.; Waterkotte, B.; Heitmann, K.; Wucherpfennig, T. Computational fluid dynamics (CFD) as a tool for industrial UF/DF tank optimization. Biochem. Eng. J. 2020, 160, 107617. [Google Scholar] [CrossRef]
- Mishra, S.; Kumar, V.; Sarkar, J.; Rathore, A.S. Mixing and mass transfer in production scale mammalian cell culture reactor using coupled CFD-species transport-PBM validation. Chem. Eng. Sci. 2023, 267, 118323. [Google Scholar] [CrossRef]
- Gelves, R.; Niño, L. CFD Prediction of Heterogeneities in the Scale up of Liquid-Liquid Dispersions. Int. J. Chem. Eng. Appl. 2014, 5, 79–84. [Google Scholar] [CrossRef]
- Kaiser, S.C.; Decaria, P.N.; Seidel, S.; Eibl, D. Scaling-up of an Insect Cell-based Virus Production Process in a Novel Single-use Bioreactor with Flexible Agitation. Chem. Ing. Tech. 2022, 94, 1950–1961. [Google Scholar] [CrossRef]
- Zhu, L.; Zhao, C.; Shiue, A.; Jeng, J.C.; Wurm, M.J.; Raussin, G.; Bürki, C.A.; Wurm, F.M. Fluid dynamics of a pilot-scale OrbShake bioreactor under different operating conditions. J. Chem. Technol. Biotechnol. 2022, 97, 1027–1036. [Google Scholar] [CrossRef]
- Cytiva. Engineering Characterization of ReadyToProcess WAVE 25 Bioreactor System with 20L Cellbag Culture Chamber. Available online: https://cdn.cytivalifesciences.com/api/public/content/digi-18120-pdf (accessed on 25 April 2024).
- Sirasitthichoke, C.; Teoman, B.; Thomas, J.; Armenante, P.M. Computational prediction of the just-suspended speed, Njs, in stirred vessels using the lattice Boltzmann method (LBM) coupled with a novel mathematical approach. Chem. Eng. Sci. 2022, 251, 117411. [Google Scholar] [CrossRef]
- Borys, B.S.; Dang, T.; So, T.; Rohani, L.; Revay, T.; Walsh, T.; Thompson, M.; Argiropoulos, B.; Rancourt, D.E.; Jung, S.; et al. Overcoming bioprocess bottlenecks in the large-scale expansion of high-quality hiPSC aggregates in vertical-wheel stirred suspension bioreactors. Stem Cell Res. Ther. 2021, 12, 55. [Google Scholar] [CrossRef] [PubMed]
- Kotidis, P.; Jedrzejewski, P.; Sou, S.N.; Sellick, C.; Polizzi, K.; del Val, I.J.; Kontoravdi, C. Model-based optimization of antibody galactosylation in CHO cell culture. Biotechnol. Bioeng. 2019, 116, 1612–1626. [Google Scholar] [CrossRef] [PubMed]
- Craven, S.; Whelan, J.; Glennon, B. Glucose concentration control of a fed-batch mammalian cell bioprocess using a nonlinear model predictive controller. J. Process Control 2014, 24, 344–357. [Google Scholar] [CrossRef]
- Grilo, A.L.; Mantalaris, A. A Predictive Mathematical Model of Cell Cycle, Metabolism, and Apoptosis of Monoclonal Antibody-Producing GS–NS0 Cells. Biotechnol. J. 2019, 14, 1800573. [Google Scholar] [CrossRef] [PubMed]
- Zürcher, P.; Sokolov, M.; Brühlmann, D.; Ducommun, R.; Stettler, M.; Souquet, J.; Jordan, M.; Broly, H.; Morbidelli, M.; Butté, A. Cell culture process metabolomics together with multivariate data analysis tools opens new routes for bioprocess development and glycosylation prediction. Biotechnol. Prog. 2020, 36, e3012. [Google Scholar] [CrossRef] [PubMed]
- Green, A.; Glassey, J. Multivariate analysis of the effect of operating conditions on hybridoma cell metabolism and glycosylation of produced antibody. J. Chem. Technol. Biotechnol. 2015, 90, 303–313. [Google Scholar] [CrossRef]
- Goldrick, S.; Lee, K.; Spencer, C.; Holmes, W.; Kuiper, M.; Turner, R.; Farid, S.S. On-Line Control of Glucose Concentration in High-Yielding Mammalian Cell Cultures Enabled Through Oxygen Transfer Rate Measurements. Biotechnol. J. 2018, 13, 1700607. [Google Scholar] [CrossRef] [PubMed]
- Kotidis, P.; Kontoravdi, C. Harnessing the potential of artificial neural networks for predicting protein glycosylation. Metab. Eng. Commun. 2020, 10, e00131. [Google Scholar] [CrossRef]
- Seidel, S.; Schirmer, C.; Maschke, R.W.; Rossi, L.; Eibl, R.; Eibl, D. Computational Fluid Dynamics for Advanced Characterisation of Bioreactors Used in the Biopharmaceutical Industry—Part I: Literature Review. In Computational Fluid Dynamics—Recent Advances, New Perspectives and Applications; IntechOpen: London, UK, 2023. [Google Scholar] [CrossRef]
- Pohar, A. A Review of Computational Fluid Dynamics (CFD) Simulations of Mixing in the Pharmaceutical Industry. Biomed. J. Sci.Tech. Res. 2020, 27, 20732–20736. [Google Scholar] [CrossRef]
- Tsopanoglou, A.; del Val, I.J. 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, 32, 100691. [Google Scholar] [CrossRef]
- Yahia, B.B.; Malphettes, L.; Heinzle, E. Macroscopic modeling of mammalian cell growth and metabolism. Appl. Microbiol. Biotechnol. 2015, 99, 7009. [Google Scholar] [CrossRef] [PubMed]
- Page, M.J.; Moher, D.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. PRISMA 2020 explanation and elaboration: Updated guidance and exemplars for reporting systematic reviews. BMJ 2021, 372, n160. [Google Scholar] [CrossRef]
- Gusenbauer, M.; Haddaway, N.R. Which academic search systems are suitable for systematic reviews or meta-analyses? Evaluating retrieval qualities of Google Scholar, PubMed, and 26 other resources. Res. Synth. Methods 2020, 11, 181–217. [Google Scholar] [CrossRef]
- Schmalzriedt, S.; Jenne, M.; Mauch, K.; Reuss, M. Integration of physiology and fluid dynamics. Adv. Biochem. Eng. Biotechnol. 2003, 80, 19–68. [Google Scholar] [CrossRef]
- Haringa, C.; Tang, W.; Deshmukh, A.T.; Xia, J.; Reuss, M.; Heijnen, J.J.; Mudde, R.F.; Noorman, H.J. Euler-Lagrange computational fluid dynamics for (bio)reactor scale down: An analysis of organism lifelines. Eng. Life Sci. 2016, 16, 652–663. [Google Scholar] [CrossRef] [PubMed]
- Morchain, J.; Gabelle, J.C.; Cockx, A. A coupled population balance model and CFD approach for the simulation of mixing issues in lab-scale and industrial bioreactors. AIChE J. 2014, 60, 27–40. [Google Scholar] [CrossRef]
- Pigou, M.; Morchain, J. Investigating the interactions between physical and biological heterogeneities in bioreactors using compartment, population balance and metabolic models. Chem. Eng. Sci. 2015, 126, 267–282. [Google Scholar] [CrossRef]
- Morchain, J.; Gabelle, J.C.; Cockx, A. Coupling of biokinetic and population balance models to account for biological heterogeneity in bioreactors. AIChE J. 2013, 59, 369–379. [Google Scholar] [CrossRef]
- Haringa, C.; Deshmukh, A.T.; Mudde, R.F.; Noorman, H.J. Euler-Lagrange analysis towards representative down-scaling of a 22 m3 aerobic S. cerevisiae fermentation. Chem. Eng. Sci. 2017, 170, 653–669. [Google Scholar] [CrossRef]
- Kuschel, M.; Siebler, F.; Takors, R. Lagrangian Trajectories to Predict the Formation of Population Heterogeneity in Large-Scale Bioreactors. Bioengineering 2017, 4, 27. [Google Scholar] [CrossRef]
- Kelly, W.J. Using computational fluid dynamics to characterize and improve bioreactor performance. Biotechnol. Appl. Biochem. 2008, 49, 225. [Google Scholar] [CrossRef]
- Haringa, C.; Noorman, H.J.; Mudde, R.F. Lagrangian modeling of hydrodynamic–kinetic interactions in (bio)chemical reactors: Practical implementation and setup guidelines. Chem. Eng. Sci. 2017, 157, 159–168. [Google Scholar] [CrossRef]
- Lapin, A.; Müller, D.; Reuss, M. Dynamic Behavior of Microbial Populations in Stirred Bioreactors Simulated with Euler−Lagrange Methods: Traveling along the Lifelines of Single Cells. Ind. Eng. Chem. Res. 2004, 43, 4647–4656. [Google Scholar] [CrossRef]
- Chen, J.; Luo, Y.; Xia, J.; Shi, Z.; Jiang, L.; Niu, S.; Zhou, X.; Cao, J. Differential responses of ecosystem respiration components to experimental warming in a meadow grassland on the Tibetan Plateau. Agric. For. Meteorol. 2016, 220, 21–29. [Google Scholar] [CrossRef]
- Yang, S.; Kiang, S.; Farzan, P.; Ierapetritou, M. Optimization of Reaction Selectivity Using CFD-Based Compartmental Modeling and Surrogate-Based Optimization. Processes 2019, 7, 9. [Google Scholar] [CrossRef]
- Zieringer, J.; Takors, R. In Silico Prediction of Large-Scale Microbial Production Performance: Constraints for Getting Proper Data-Driven Models. Comput. Struct. Biotechnol. J. 2018, 16, 246–256. [Google Scholar] [CrossRef]
- Clincke, M.F.; Mölleryd, C.; Samani, P.K.; Lindskog, E.; Fäldt, E.; Walsh, K.; Chotteau, V. Very High Density of Chinese Hamster Ovary Cells in Perfusion by Alternating Tangential Flow or Tangential Flow Filtration in WAVE BioreactorTM—Part II: Applications for Antibody Production and Cryopreservation. Biotechnol. Prog. 2013, 29, 768. [Google Scholar] [CrossRef] [PubMed]
- Lapin, A.; Schmid, J.; Reuss, M. Modeling the dynamics of E. coli populations in the three-dimensional turbulent field of a stirred-tank bioreactor—A structured–segregated approach. Chem. Eng. Sci. 2006, 61, 4783–4797. [Google Scholar] [CrossRef]
- Raffel, M.; Willert, C.E.; Scarano, F.; Kähler, C.J.; Wereley, S.T.; Kompenhans, J. Particle Image Velocimetry: A Practical Guide, 3rd ed.; Springer: Berlin/Heidelberg, Germany, 2018. [Google Scholar] [CrossRef]
- Larsson, G.; Törnkvist, M.; Wernersson, S.E.; Trägårdh, C.; Noorman, H.; Enfors, S.O. Substrate gradients in bioreactors: Origin and consequences. Bioprocess Eng. 1996, 14, 281–289. [Google Scholar] [CrossRef]
- Barbouche, N.; Olmos, E.; Guedon, E.; Marc, A. Coupling between Cell Kinetics and CFD to Establish Physio-Hydrodynamic Correlations in Various Stirred Culture Systems. In Proceedings of the Cells and Culture: Proceedings of the 20th ESACT Meeting, Dresden, Germany, 17–20 June 2007; pp. 213–217. [Google Scholar] [CrossRef]
- Nadal-Rey, G.; Kavanagh, J.M.; Cassells, B.; Cornelissen, S.; Fletcher, D.F.; Gernaey, K.V.; McClure, D.D. Modelling of industrial-scale bioreactors using the particle lifeline approach. Biochem. Eng. J. 2023, 198, 108989. [Google Scholar] [CrossRef]
- Oliveira, C.L.; Pace, Z.; Thomas, J.A.; DeVincentis, B.; Sirasitthichoke, C.; Egan, S.; Lee, J. CFD-based bioreactor model with proportional-integral-derivative controller functionality for dissolved oxygen and pH. Biotechnol. Bioeng. 2024, 121, 655–669. [Google Scholar] [CrossRef] [PubMed]
- Hanspal, N.; DeVincentis, B.; Thomas, J.A. Modeling multiphase fluid flow, mass transfer, and chemical reactions in bioreactors using large-eddy simulation. Eng. Life Sci. 2023, 23, e2200020. [Google Scholar] [CrossRef] [PubMed]
- Lao-Martil, D.; Schmitz, J.P.J.; Teusink, B.; van Riel, N.A.W. Elucidating yeast glycolytic dynamics at steady state growth and glucose pulses through kinetic metabolic modeling. Metab. Eng. 2023, 77, 128–142. [Google Scholar] [CrossRef] [PubMed]
- Rojek, K.; Wyrzykowski, R.; Gepner, P. AI-Accelerated CFD Simulation Based on OpenFOAM and CPU/GPU Computing; Springer: Berlin/Heidelberg, Germany, 2021; pp. 373–385. [Google Scholar] [CrossRef]
- Blöbaum, L.; Haringa, C.; Grünberger, A. Microbial lifelines in bioprocesses: From concept to application. Biotechnol. Adv. 2023, 62, 108071. [Google Scholar] [CrossRef] [PubMed]
- Haringa, C.; Tang, W.; Noorman, H.J. Stochastic parcel tracking in an Euler–Lagrange compartment model for fast simulation of fermentation processes. Biotechnol. Bioeng. 2022, 119, 1849–1860. [Google Scholar] [CrossRef] [PubMed]
- Spann, R.; Glibstrup, J.; Pellicer-Alborch, K.; Junne, S.; Neubauer, P.; Roca, C.; Kold, D.; Lantz, A.E.; Sin, G.; Gernaey, K.V. CFD predicted pH gradients in lactic acid bacteria cultivations. Biotechnol. Bioeng. 2019, 116, 769–780. [Google Scholar] [CrossRef] [PubMed]
- Nașcu, I.; Sebastia-Saez, D.; Chen, T.; Nașcu, I.; Du, W. Global sensitivity analysis for a perfusion bioreactor based on CFD modelling. Comput. Chem. Eng. 2022, 163, 107829. [Google Scholar] [CrossRef]
- Farzan, P.; Ierapetritou, M.G. Integrated modeling to capture the interaction of physiology and fluid dynamics in biopharmaceutical bioreactors. Comput. Chem. Eng. 2017, 97, 271–282. [Google Scholar] [CrossRef]
- Tang, W.; Deshmukh, A.T.; Haringa, C.; Wang, G.; Gulik, V.W.; Winden, V.W.; Reuss, M.; Heijnen, J.J.; Xia, J.; Chu, J. A 9-pool metabolic structured kinetic model describing days to seconds dynamics of growth and product formation by Penicillium chrysogenum. Biotechnol. Bioeng. 2017, 114, 1733–1743. [Google Scholar] [CrossRef]
- Haringa, C.; Tang, W.; Wang, G.; Deshmukh, A.T.; van Winden, W.A.; Chu, J.; van Gulik, W.M.; Heijnen, J.J.; Mudde, R.F.; Noorman, H.J. Computational fluid dynamics simulation of an industrial P. chrysogenum fermentation with a coupled 9-pool metabolic model: Towards rational scale-down and design optimization. Chem. Eng. Sci. 2018, 175, 12–24. [Google Scholar] [CrossRef]
- Liu, Y.; Wang, Z.J.; Xia, J.Y.; Haringa, C.; Liu, Y.P.; Chu, J.; Zhuang, Y.P.; Zhang, S.L. Application of Euler–Lagrange CFD for quantitative evaluating the effect of shear force on Carthamus tinctorius L. cell in a stirred tank bioreactor. Biochem. Eng. J. 2016, 114, 209–217. [Google Scholar] [CrossRef]
- McClure, D.D.; Kavanagh, J.M.; Fletcher, D.F.; Barton, G.W.; Al, E. Characterizing bubble column bioreactor performance using computational fluid dynamics. Chem. Eng. Sci. 2016, 144, 58–74. [Google Scholar] [CrossRef]
- Sonnleitner, B.; Käppeli, O. Growth of Saccharomyces cerevisiae is controlled by its limited respiratory capacity: Formulation and verification of a hypothesis. Biotechnol. Bioeng. 1986, 28, 927–937. [Google Scholar] [CrossRef]
- Mcclure, D.D.; Kavanagh, J.M.; Fletcher, D.F.; Barton, G.W. Development of a CFD Model of Bubble Column Bioreactors: Part Two—Comparison of Experimental Data and CFD Predictions. Chem. Eng. Technol. 2014, 37, 131–140. [Google Scholar] [CrossRef]
- Xu, B.; Jahic, M.; Enfors, S.O. Modeling of overflow metabolism in batch and fed-batch cultures of Escherichia coli. Biotechnol. Prog. 1999, 15, 81–90. [Google Scholar] [CrossRef]
- Delafosse, A.; Line, A.; Morchain, J.; Guiraud, P. LES and URANS simulations of hydrodynamics in mixing tank: Comparison to PIV experiments. Chem. Eng. Res. Des. 2008, 86, 1322–1330. [Google Scholar] [CrossRef]
- Gabelle, J.C.; Augier, F.; Carvalho, A.; Rousset, R.; Morchain, J. Effect of tank size on kLa and mixing time in aerated stirred reactors with non-newtonian fluids. Can. J. Chem. Eng. 2011, 89, 1139–1153. [Google Scholar] [CrossRef]
- Kätterer, L.; Allemann, H.; Käppeli, O.; Fiechter, A. Transient responses of continuously growing yeast cultures to dilution rate shifts: A sensitive means to analyze biology and the performance of equipment. Biotechnol. Bioeng. 1986, 28, 146–150. [Google Scholar] [CrossRef] [PubMed]
- Elqotbi, M.; Vlaev, S.D.; Montastruc, L.; Nikov, I. CFD modelling of two-phase stirred bioreaction systems by segregated solution of the Euler-Euler model. Comput. Chem. Eng. 2013, 48, 113–120. [Google Scholar] [CrossRef]
- Zhang, J.; Geng, X. Numerical simulation of proteins flowing through a bioreactor coupled with biochemical reactions on the boundary. Int. J. Nonlinear Sci. Numer. Simul. 2010, 11, 1003–1010. [Google Scholar] [CrossRef]
- Jenne, M.; Reuss, M. A critical assessment on the use of k-ε turbulence models for simulation of the turbulent liquid flow induced by a Rushton-turbine in baffled stirred-tank reactors. Chem. Eng. Sci. 1999, 54, 3921–3941. [Google Scholar] [CrossRef]
- Chassagnole, C.; Noisommit-Rizzi, N.; Schmid, J.W.; Mauch, K.; Reuss, M. Dynamic modeling of the central carbon metabolism of Escherichia coli. Biotechnol. Bioeng. 2002, 79, 53–73. [Google Scholar] [CrossRef] [PubMed]
- Narayanan, H.; Sokolov, M.; Morbidelli, M.; Butté, A. A new generation of predictive models: The added value of hybrid models for manufacturing processes of therapeutic proteins. Biotechnol. Bioeng. 2019, 116, 2540–2549. [Google Scholar] [CrossRef] [PubMed]
- Delafosse, A.; Calvo, S.; Collignon, M.L.; Delvigne, F.; Crine, M.; Toye, D. Euler-Lagrange approach to model heterogeneities in stirred tank bioreactors—Comparison to experimental flow characterization and particle tracking. Chem. Eng. Sci. 2015, 134, 457–466. [Google Scholar] [CrossRef]
- Pirker, S.; Lichtenegger, T. Efficient time-extrapolation of single- and multiphase simulations by transport based recurrence CFD (rCFD). Chem. Eng. Sci. 2018, 188, 65–83. [Google Scholar] [CrossRef]
Database | CFD Modelling | CRK Modelling | System | Limit to | ||
---|---|---|---|---|---|---|
Scopus | “Computational Fluid Dynamics” OR “CFD” | AND | “Cell Reaction Kinetics” OR “Kinetic modelling” | AND | “Fermentation” OR “Bioreactor” OR “Bioprocess” OR “Bioprocess development” | Source Type: Journals Document Type: Articles, Conference papers Language: English |
Factors | CFD Approach | Compartmentalisation-Based Modelling Approach | |
---|---|---|---|
Eulerian Approach | Agent-Based Methodology | ||
Computational effort | High | High | Low |
Level of accuracy | High | High | Low |
Prediction accuracy of flow regime | High | High | Low |
Single-cell tracking | No | Yes | Yes |
Integrable model size | Coarse-grained small-scale | Coarse-grained small-scale | Genome-scale |
Number of particles | High | Low (<10%) | High |
Bioreactor Physical Conditions | CFD-CRK Coupling Strategy | Computational Load | Model Prediction Accuracy | Remarks |
---|---|---|---|---|
Small-scale bioreactor with heterogenous culture parameters in spacetime | Eulerian integration | Low | Low | The application of Eulerian integration to a heterogeneous bioprocessing environment is an oversimplification of the system, which leads to poor prediction of culture parameters [76]. This coupling strategy can be used to integrate complex and unstable cellular models to formulate the sample space for the process conditions to be further evaluated. |
Small-scale bioreactor with temporally heterogeneous but spatially homogeneous culture parameters | Eulerian integration | Low | Medium | The assumption of spatially homogeneous culture parameters represents an ideally mixed system, which can be loosely approximated to be the scenario in small-volume bioreactors with ample agitation and aeration. In such cases, model prediction is postulated to increase as cellular behaviour is only temporally impacted. |
Small-scale bioreactor with homogeneous culture parameters in spacetime | Eulerian integration | Low | High | The assumption of homogeneous culture parameters in spacetime is a hypothetical case. The closest example of such a case is the production phase of a small-scale, continuously perfused cell culture process, as the nutrient availability and distribution happen in a close to uniform environment. For this duration of steady state, Eulerian coupling can provide higher prediction accuracy. |
Small-scale bioreactor with heterogenous culture parameters in spacetime | Phase ensemble average-based Lagrangian integration | Medium | Medium | Phase ensemble average-based Lagrangian integration ignores the spatial heterogeneity of the parameters and uses a time-averaged approach to account for the temporal variation of culture parameters. This increases the computational time as well as the accuracy of the predicted model compared to the Eulerian approach, which completely ignores the presence of heterogeneous culture parameters in spacetime. |
Small-scale bioreactor with heterogenous culture parameters in spacetime | Grid cell average-based Lagrangian | High | High | Grid cell average-based Lagrangian approach tracks the cells for the variations in cell behaviour in spacetime. Clearly, this approach is computationally expensive but offers high-quality resolution in terms of prediction accuracy. Such a coupling approach can be applied to small-scale bioreactors with reduced cellular models. |
Small-scale bioreactor with heterogenous culture parameters in spacetime | Multizonal grid cell average-based Lagrangian | Medium to High | Medium to High | The multizonal grid cell average-based Lagrangian approach divides the bioreactor 3D space into multiple compartments and assumes spatial uniformity within these zones. This approach reduces the computational burden as compared to the non-compartmentalised grid cell average-based Lagrangian approach, and the appropriate selection of multizone ensures medium to high prediction accuracy. |
Large-scale bioreactor with heterogenous culture parameters in spacetime | Eulerian integration | Low | Low | For large-scale bioreactors, CFD models are less computationally tractable, thereby increasing the simulation time [78]. Eulerian coupling is the only feasible approach currently, and technological advancements in computing are required to move to better coupling approaches capable of providing more realistic prediction accuracy. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Singh, V.K.; Jiménez del Val, I.; Glassey, J.; Kavousi, F. Integration Approaches to Model Bioreactor Hydrodynamics and Cellular Kinetics for Advancing Bioprocess Optimisation. Bioengineering 2024, 11, 546. https://doi.org/10.3390/bioengineering11060546
Singh VK, Jiménez del Val I, Glassey J, Kavousi F. Integration Approaches to Model Bioreactor Hydrodynamics and Cellular Kinetics for Advancing Bioprocess Optimisation. Bioengineering. 2024; 11(6):546. https://doi.org/10.3390/bioengineering11060546
Chicago/Turabian StyleSingh, Vishal Kumar, Ioscani Jiménez del Val, Jarka Glassey, and Fatemeh Kavousi. 2024. "Integration Approaches to Model Bioreactor Hydrodynamics and Cellular Kinetics for Advancing Bioprocess Optimisation" Bioengineering 11, no. 6: 546. https://doi.org/10.3390/bioengineering11060546
APA StyleSingh, V. K., Jiménez del Val, I., Glassey, J., & Kavousi, F. (2024). Integration Approaches to Model Bioreactor Hydrodynamics and Cellular Kinetics for Advancing Bioprocess Optimisation. Bioengineering, 11(6), 546. https://doi.org/10.3390/bioengineering11060546