Artificial intelligence and machine learning applications in biopharmaceutical manufacturing

Artificial intelligence and machine learning applications in biopharmaceutical manufacturing

Source Node: 1885256
    • Charniak Eugene

    Introduction to Artificial Intelligence.

    Pearson Education India, 1985

    • Copeland Jack

    Artificial Intelligence: A Philosophical Introduction.

    John Wiley & Sons, 2015

    • Jordan M.I.
    • Mitchell T.M.

    Machine learning: trends, perspectives, and prospects.

    Science. 2015; 349: 255-260

    • MarketsandMarkets

    Artificial intelligence in healthcare market by offering (hardware, software, services), technology (machine learning, NLP, context-aware computing, end-use application, end user, and geography: Global forecast to 2025.

    () MarketsandMarkets, 2018

    • Mak Kit-Kay
    • et al.

    Artificial intelligence in drug development: present status and future prospects.

    Drug Discov. Today. 2019; 24: 773-780

    • Fleming Nic

    How artificial intelligence is changing drug discovery.

    Nature. 2018; 557: S55-S57

    • Viceconti Marco
    • et al.

    In silico clinical trials: how computer simulation will transform the biomedical industry.

    Int. J. Clin. Trials. 2016; 3: 37-46

  • An AI boost for clinical trials.

    Nature. 2019; 573: S100-S102

    • Harrer Stefan
    • et al.

    Artificial intelligence for clinical trial design.

    Trends Pharmacol. Sci. 2019; 40: 577-591

    • Opderbeck David W.

    Artificial Intelligence in pharmaceuticals, biologics, and medical devices: present and future regulatory models.

    Fordham L. Rev. 2019; 88: 553

    • Ramkumar Prem N.
    • et al.

    Artificial intelligence and arthroplasty at a single institution: real-world applications of machine learning to big data, value-based care, mobile health, and remote patient monitoring.

    J. Arthroplast. 2019; 34: 2204-2209

    • Nargund Shilpa
    • et al.

    The move toward biopharma 4.0: in silico biotechnology develops “smart” processes that benefit biomanufacturing through Digital Twins.

    Genet. Eng. Biotechnol. News. 2019; 39: 53-55

    • Li Bh.
    • et al.

    Applications of artificial intelligence in intelligent manufacturing: a review.

    Front. Inf. Technol. Electron. Eng. 2017; 18: 86-96

    • Steedman M.
    • et al.

    Intelligent biopharma – forging the links across the value chain, Deloitte Insights.

    2019 ()

    • Sertkaya Aylin
    • et al.

    Key cost drivers of pharmaceutical clinical trials in the United States.

    Clin. Trials. 2016; 13: 117-126

    • Wuest Thorsten
    • et al.

    Machine learning in manufacturing: advantages, challenges, and applications.

    Prod. Manufact. Res. 2016; 4: 23-45

  • PAT — A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality Assurance.

    • Narayanan Harini
    • et al.

    Hybrid Models for the simulation and prediction of chromatographic processes for protein capture.

    J. Chromatogr. A. 2021; 1650462248https://doi.org/10.1016/j.chroma.2021.462248

    • von Stosch M.
    • et al.

    Hybrid modeling for quality by design and PAT-benefits and challenges of applications in biopharmaceutical industry.

    Biotechnol. J. 2014; 9: 719-726

    • Bayer B.
    • et al.

    Hybrid modeling and intensified DoE: an approach to accelerate upstream process characterization.

    Biotechnol. J. 2020; 15: 2000121

    • Bayer B.
    • et al.

    Digital twin application for model-based DoE to rapidly identify ideal process conditions for space-time yield optimization.

    Processes. 2021; 9: 1109

    • Krippl M.
    • et al.

    Hybrid modeling for simultaneous prediction of flux, rejection factor and concentration in two-component crossflow ultrafiltration.

    Processes. 2020; 8: 1625

    • Glassey Jarka
    • et al.

    Process analytical technology (PAT) for biopharmaceuticals.

    Biotechnol. J. 2011; 6: 369

    • Rathore Anurag S.
    • et al.

    Guidance for performing multivariate data analysis of bioprocessing data: pitfalls and recommendations.

    Biotechnol. Prog. 2014; 30: 967-973

    • Smiatek J.
    • et al.

    Towards a digital bioprocess replica: computational approaches in biopharmaceutical development and manufacturing.

    Trends Biotechnol. 2020; 38: 1141-1153

    • Zheng H.
    • et al.

    Green simulation assisted reinforcement learning with model risk for biomanufacturing learning and control.

    2006 ()

    • Nikita S.
    • et al.

    Reinforcement learning based optimization of process chromatography for continuous processing of biopharmaceuticals.

    Chem. Eng. Sci. 2021; 230116171

    • Canzoneri Michelangelo
    • et al.

    Digital Twins: A General Overview of the Biopharma Industry.

    Adv. Biochem. Eng. Biotechnol. 2021; 2021: 167-184

    • Baughman D.R.
    • Liu Y.A.

    Neural Networks In Bioprocessing And Chemical Engineering.

    Academic Press, 1995

    • Bishop Christopher M.

    Pattern Recognition and Machine Learning.

    Springer, 2006

    • Norvig Peter
    • Russell Stuart J.

    Artificial Intelligence: A Modern Approach.

    Pearson, 2010

  • Evolving artificial neural networks.

    Proc. IEEE. 1999; 87: 1423-1447

    • Graupe Daniel

    Principles of Artificial Neural Networks.

    Vol. 7. World Scientific, 2013

    • Karayiannis Nicolaos
    • et al.

    Artificial Neural Networks: Learning Algorithms, Performance Evaluation, and Applications.

    Vol. 209. Springer Science & Business Media, 1992

    • Kaelbling Leslie Pack
    • et al.

    Reinforcement learning: a survey.

    J. Artif. Intell. Res. 1996; 4: 237-285

    • Silver David
    • et al.

    Mastering the game of go without human knowledge.

    Nature. 2017; 550: 354-359

  • Deep Reinforcement Learning Hands-On: Apply Modern RL Methods, with Deep Q-networks, Value Iteration, Policy Gradients, TRPO, AlphaGo Zero and More.

    Packt Publishing Ltd, 2018

    • Thrun Sebastian B.

    Efficient Exploration in Reinforcement Learning. Technical Report.

    Carnegie Mellon University, 1992

    • Ozturk Sadettin S.

    Engineering challenges in high density cell culture systems.

    Cytotechnology. 1996; 22: 3-16

    • Luchner M.
    • et al.

    Online prediction of product titer and solubility of recombinant proteins in Escherichia coli fed-batch cultivations.

    J. Chem. Technol. Biotechnol. 2015; 90: 283-290

    • Melcher M.
    • et al.

    The potential of random forest and neural networks for biomass and recombinant protein modeling in Escherichia coli fed-batch fermentations.

    Biotechnol. J. 2015; 10: 1770-1782

    • Chiappini A.F.
    • et al.

    Modeling of bioprocess non-linear fluorescence data for at-line prediction of etanercept based on artificial neural networks optimized by response surface methodology.

    Talanta. 2020; 210120664

    • Takahashi M.B.
    • et al.

    Artificial neural network associated to UV/Vis spectroscopy for monitoring bioreactions in biopharmaceutical processes.

    Bioprocess Biosyst. Eng. 2015; 38: 1045-1054

    • Hosseini S.N.
    • et al.

    Accurate and cost-effective prediction of HBsAg titer in industrial scale fermentation process of recombinant Pichia pastoris by using neural network based soft sensor.

    Biotechnol. Appl. Biochem. 2019; 66: 681-689

    • Akisue A.R.
    • et al.

    Development of a fuzzy system for dissolved oxygen control in a recombinant Escherichia coli cultivation for heterologous protein expression.

    Comput. Aided Chem. Eng. 2018; 43: 1129-1134

    • Beiroti A.
    • et al.

    Comparative study of m-stat methanol feeding control in fed-batch fermentation of Pichia pastoris producing HBsAg: an open-loop control versus recurrent artificial neural network-based feedback control.

    J. Chem. Technol. Biotechnol. 2019; 94: 3924-3931

    • Natarajan P.
    • et al.

    Online deep neural network-based feedback control of a lutein bioprocess.

    J. Process Control. 2021; 98: 41-51

    • von Stosch M.
    • et al.

    Hybrid modeling as a QbD/PAT tool in process development: an industrial E. coli case study.

    Bioprocess Biosyst. Eng. 2016; 39: 773-784

    • Durand-P O.
    • et al.

    Artificial neural network for bioprocess monitoring based on fluorescence measurements: training without offline measurements.

    Eng. Life Sci. 2017; 17: 874-880

    • Chen Hongming
    • et al.

    The rise of deep learning in drug discovery.

    Drug Discov. Today. 2018; 23: 1241-1250

    • Popova Mariya
    • et al.

    Deep reinforcement learning for de novo drug design.

    Sci. Adv. 2018; 4eaap7885

    • Zhou Zhenpeng
    • et al.

    Optimization of molecules via deep reinforcement learning.

    Sci. Rep. 2019; 9: 1-10

    • Treloar N.J.
    • et al.

    Deep reinforcement learning for the control of microbial co-cultures in bioreactors.

    PLoS Comput. Biol. 2020; 16e1007783

    • Wang B.
    • et al.

    Reinforcement learning under model risk for biomanufacturing fermentation control.

    2021 ()

    • Karst Daniel Johannes
    • et al.

    Continuous integrated manufacturing of therapeutic proteins.

    Curr. Opin. Biotechnol. 2018; 53: 76-84

    • Kroll Paul
    • et al.

    Model-based methods in the biopharmaceutical process lifecycle.

    Pharm. Res. 2017; 34: 2596-2613

    • Luttmann Reiner
    • et al.

    Soft sensors in bioprocessing: a status report and recommendations.

    7. 2012: 1040-1048

    • Ramin Pedram
    • et al.

    Modelling continuous pharmaceutical and bio-based processes at plant-wide level: a roadmap towards efficient decision-making.

    Chem. Today. 2018; 36: 26-30

    • Zalai Denes
    • et al.

    Microbial technologies for biotherapeutics production: key tools for advanced biopharmaceutical process development and control.

    Drug Discov. Today Technol. 2020; 38: 9-24

    • Wang G.
    • et al.

    Root cause investigation of deviations in protein chromatography based on mechanistic models and artificial neural networks.

    J. Chromatogr. A. 2017; 1515: 146-153

  • D.Q. Lin, et al. (2019) Design and analysis of multi-column continuous chromatography based on artificial neural network. China Patent CN110348090A

    • Agarwal H.
    • et al.

    Artificial neural network (ANN)-based prediction of depth filter loading capacity for filter sizing.

    Biotechnol. Prog. 2016; 32: 1436-1443

    • Krippl Maximilian
    • et al.

    Hybrid modeling of cross-flow filtration: predicting the flux evolution and duration of ultrafiltration processes.

    Sep. Purif. Technol. 2020; 248117064

    • Krippl Maximilian
    • et al.

    Hybrid modeling for simultaneous prediction of flux, rejection factor and concentration in two-component crossflow ultrafiltration.

    Processes. 2020; 8: 1625

    • Garcel R.H.R.
    • et al.

    Preliminary modeling of an industrial recombinant human erythropoietin purification process by artificial neural networks.

    Braz. J. Chem. Eng. 2015; 32: 725-734

    • Pirrung S.M.
    • et al.

    Optimization of biopharmaceutical downstream processes supported by mechanistic models and artificial neural networks.

    Biotechnol. Prog. 2017; 33: 696-707

    • Pirrung S.M.
    • et al.

    Model-based optimization of integrated purification sequences for biopharmaceuticals.

    Chem. Eng. Sci. X. 2019; 3100025

    • Li Y.F.
    • et al.

    Neural network to understand process capability and process intermediates acceptance criteria in monoclonal antibody production process.

    J. Pharm. Innov. 2018; 13: 36-47

    • Fahey W.
    • et al.

    Improving biopharmaceutical manufacturing yield using neural network classification.

    Bioprocess. J. 2016; 14: 39-50

    • Hwangbo S.
    • et al.

    Design of control framework based on deep reinforcement learning and Monte-Carlo sampling in downstream separation.

    Comput. Chem. Eng. 2020; 140106910

    • Hwangbo S.
    • et al.

    Design of smart liquid-liquid extraction columns for downstream separations of biopharmaceuticals using deep Q-learning algorithm.

    in: Computer Aided Chemical Engineering. Elsevier, 2019: 271-276

    • Badillo S.
    • et al.

    An introduction to machine learning.

    Clin. Pharmacol. Ther. 2020; 107: 871-885

    • Sutton Richard S.
    • et al.

    Reinforcement Learning: An Introduction.

    MIT Press, 2018

    • Smola A.J.
    • Vishwanathan S.V.N.

    Introduction to Machine Learning.

    Vol. 32. Cambridge University, 2008: 2008

    • Alpaydin Ethem

    Introduction to Machine Learning.

    MIT press, 2020

    • Ciaburro Giuseppe
    • et al.

    Neural Networks With R: Smart Models Using CNN, RNN, Deep Learning, and Artificial Intelligence Principles.

    Packt Publishing Ltd, 2017: 2017

    • Paluszek Michael
    • et al.

    MATLAB Machine Learning.

    Apress, 2016: 2016

    • Garcia-Ochoa Felix
    • et al.

    Bioreactor scale-up and oxygen transfer rate in microbial processes: an overview.

    Biotechnol. Adv. 2009; 27: 153-176

    • Otto Ralf
    • et al.

    Rapid Growth in Biopharma: Challenges and Opportunities.

    McKinsey & Company, 2014

    • Rathore Anurag S.
    • et al.

    Quality by design for biopharmaceuticals.

    Nat. Biotechnol. 2009; 27: 26-34

    • Subramanian Ganapathy

    Continuous Processing in Pharmaceutical Manufacturing.

    John Wiley & Sons, 2014

    • Ündey Cenk
    • et al.

    Applied advanced process analytics in biopharmaceutical manufacturing: challenges and prospects in real-time monitoring and control.

    J. Process Control. 2010; 20: 1009-1018

    • Thakur Garima
    • et al.

    Control of surge tanks for continuous manufacturing of monoclonal antibodies.

    Biotechnol. Bioeng. 2021; 118: 1913-1931

    • Zeberli Anicia
    • et al.

    Data-driven anomaly detection and diagnostics for changeover processes in biopharmaceutical drug product manufacturing.

    Chem. Eng. Res. Des. 2021; 167: 53-62

    • Borchert D.
    • et al.

    Comparison of data science workflows for root cause analysis of bioprocesses.

    Bioprocess Biosyst. Eng. 2018; 42: 245-256

    • Hebbi Vishwanath
    • et al.

    Process analytical technology implementation for protein refolding: GCSF as a case study.

    Biotechnol. Bioeng. 2019; 116: 1039-1052

    • Sachs L.
    • Hedderich J.

    Angewandte Statistik. Methodensammlung mit R.

    Springer, 2006

    • Hyndman R.J.
    • Koehler A.B.

    Another look at measures of forecast accuracy.

    Int. J. Forecast. 2006; 22: 679-688

  • Basic principles of ROC analysis.

    Semin. Nucl. Med. 1978; 8: 283-298

    • Olson D.L.
    • Delen D.

    Advanced Data Mining Techniques.

    Springer Publishing, 2008

  • Guidance for Industry, Q2B Validation of Analytical Procedures: Methodology; FDA-1996-D-0169.

    Center for Drug Evaluation and Research, 1997

    • Guo S.
    • et al.

    Common mistakes in cross-validating classification models.

    Anal. Methods. 2017; 9: 4410-4417

    • Quiroz J.
    • Burdick R.K.

    Assessing equivalence of two assays using sensitivity and specificity.

    J. Biopharm. Stat. 2007; 17: 433-443

    • Brereton R.G.
    • et al.

    Chemometrics in analytical chemistry—Part II: modeling, validation, and applications.

    Anal. Bioanal. Chem. 2018; 410: 6691-6704

    • Westad F.
    • Marini F.

    (2015) Validation of chemometric models—a tutorial.

    Anal. Chim. Acta. 2015; 893: 14-24

  • ICH Q2 (R1) Validation of Analytical Procedures: Text and Methodology.

    ICH, 1997

  • Evaluation of intermediate precision in the validation of analytical procedures for drugs: from NDA dossiers.

    Pharm. Tech. Jpn. 2002; 18: 695-704

  • Nonparametric maximum likelihood estimation of a mixing distribution.

    J. Am. Stat. Assoc. 1978; 73: 805-811

    • Cavanaugh J.E.
    • Neath A.A.

    The Akaike information criterion: background, derivation, properties, application, interpretation, and refinements.

    WIREs Comput. Stat. 2019; 11e1460

    • Maydeu-Olivares A.
    • García-Forero C.

    Goodness-of-Fit. Testing.

    in: Peterson P. Baker E. McGaw B. International Encyclopedia of Education. 3rd ed. Elsevier, 2010: 190-196

    • Liu Y.
    • Gunawan R.

    (2017) Bioprocess optimization under uncertainty using ensemble modeling.

    J. Biotechnol. 2017; 244: 34-44

    • Viceconti M.
    • et al.

    In silico trials: verification, validation and uncertainty quantification of predictive models used in the regulatory evaluation of biomedical products.

    Methods. 2021; 185: 120-127

    • Bideault G.
    • et al.

    Verification and validation of computational models used in biopharmaceutical manufacturing: potential application of the ASME verification and validation 40 standard and FDA proposed AI/ML model. Life cycle management framework.

    J. Pharm. Sci. 2021; 110: 1540-1544

    • Smiatek J.
    • et al.

    Validation is not verification: precise terminology and scientific methods in bioprocess modeling.

    Trends Biotechnol. 2021; 39: 1117-1119

    • Der Kiureghian Armen
    • Ditlevsen Ove

    Aleatory or epistemic? Does it matter?.

    Struct. Saf. 2009; 31: 105-112

    • Abdar Moloud
    • et al.

    A review of uncertainty quantification in deep learning: Techniques, applications and challenges.

    Inform. Fusion. 2021; 76: 243-297

    • Lin X.
    • et al.

    A method for handling metabonomics data from liquid chromatography/mass spectrometry: combinational use of support vector machine recursive feature elimination, genetic algorithm and random forest for feature selection.

    Metabolomics. 2011; 7: 549-558

    • Ren J.
    • et al.

    Application of a kNN-based similarity method to biopharmaceutical manufacturing.

    Biotechnol. Prog. 2020; 36e2945

    • Szabó É.
    • et al.

    Linear discriminant analysis, partial least squares discriminant analysis, and soft independent modeling of class analogy of experimental and simulated near-infrared spectra of a cultivation medium for mammalian cells.

    J. Chemom. 2018; 32e3005

    • Casola G.
    • et al.

    Data mining algorithm for pre-processing biopharmaceutical drug product manufacturing records.

    Comput. Chem. Eng. 2019; 124: 253-269

    • Narayanan H.
    • et al.

    Decision Tree-PLS (DT-PLS) algorithm for the development of process: specific local prediction models.

    Biotechnol. Prog. 2019; 35e2818

    • Schaub J.
    • et al.

    Advancing biopharmaceutical process development by system-level data analysis and integration of omics data.

    in: Hu W. Zeng A.P. Genomics and Systems Biology of Mammalian Cell Culture. vol 127. Springer, Berlin, Heidelberg2011

    • Cheema J.J.
    • et al.

    Genetic programming assisted stochastic optimization strategies for optimization of glucose to gluconic acid fermentation.

    Biotechnol. Prog. 2002; 18: 1356-1365

    • Prabhu A.A.
    • et al.

    Biovalorisation of crude glycerol and xylose into xylitol by oleaginous yeast Yarrowia lipolytica.

    Microb. Cell Factories. 2020; 19: 121

  • Time Stamp:

    More from Biotechnology Trends