Skip to main content
Log in

Model-assisted Design of Experiments as a concept for knowledge-based bioprocess development

  • Research Paper
  • Published:
Bioprocess and Biosystems Engineering Aims and scope Submit manuscript

Abstract

Design of Experiments methods offer systematic tools for bioprocess development in Quality by Design, but their major drawback is the user-defined choice of factor boundary values. This can lead to several iterative rounds of time-consuming and costly experiments. In this study, a model-assisted Design of Experiments concept is introduced for the knowledge-based reduction of boundary values. First, the parameters of a mathematical process model are estimated. Second, the investigated factor combinations are simulated instead of experimentally derived and a constraint-based evaluation and optimization of the experimental space can be performed. The concept is discussed for the optimization of an antibody-producing Chinese hamster ovary batch and bolus fed-batch process. The same optimal process strategies were found if comparing the model-assisted Design of Experiments (4 experiments each) and traditional Design of Experiments (16 experiments for batch and 29 experiments for fed-batch). This approach significantly reduces the number of experiments needed for knowledge-based bioprocess development.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Abbreviations

\(\alpha \) :

Constant antibody production rate (\(\hbox {mg} \, \hbox {cell}^{-1} \, \hbox {h}^{-1}\))

\(\mu \) :

Cell-specific growth rate (\(\hbox {h}^{-1}\))

\(\mu _{\mathrm{d,max}} \) :

Maximum death rate (\(\hbox {h}^{-1}\))

\(\mu _{\mathrm{d,min}} \) :

Minimum death rate (\(\hbox {h}^{-1}\))

\(\mu _{\mathrm{max}} \) :

Maximum growth rate (\(\hbox {h}^{-1}\))

\(c_{{i}} \) :

Concentration of component i (\(\hbox {mmol} \, \hbox {l}^{-1}\))

\(d_{{i}} \) :

Desirability function (−)

D :

Overall desirability function (−)

\(F_{{i}} \) :

Feed concentration of component i (\(\hbox {mmol} \, \hbox {l}^{-1}\))

\(F_{\mathrm{rate}} \) :

Feed rate (\(\hbox {ml} \, \hbox {d}^{-1}\))

\(\hbox {Feed-start}\) :

Time of feed-start (\(\hbox {h}\))

i :

Index (Glc, Gln, Amm, Lac, mAb) (−)

j :

Index (lactate, ammonium)

\(k_{{i}}\) :

Inhibitory constant (\(\hbox {mmol} \, \hbox {l}^{-1}\))

\(K_{\mathrm{i,Amm}}\) :

Inhibitory constant of ammonia (\(\hbox {mmol} \, \hbox {l}^{-1}\))

\(K_{\mathrm{Lys}}\) :

Cell lysis constant (\(\hbox {h}^{-1}\))

\(K_{\mathrm{S,i}}\) :

Monod kinetic constant for component i (\(\hbox {mmol} \, \hbox {l}^{-1}\))

\(L_{{i}}\) :

Lower acceptable response (−)

\(q_{\mathrm{Amm}}\) :

Ammonia formation rate (\(\hbox {mmol} \, \hbox {cell}^{-1} \, \hbox {h}^{-1}\))

\(q_{\mathrm{Glc}} \) :

Glucose uptake rate (\(\hbox {mmol} \, \hbox {cell}^{-1} \, \hbox {h}^{-1}\))

\(q_{\mathrm{Gln}} \) :

Glutamine uptake rate (\(\hbox {mmol} \, \hbox {cell}^{-1} \, \hbox {h}^{-1}\))

\(q_{i,\mathrm{max}} \) :

Maximum uptake rate (component i) (\(\hbox {mmol} \, \hbox {cell}^{-1} \, \hbox {h}^{-1}\))

\(q_{\mathrm{Lac}} \) :

Lactate formation rate (\(\hbox {mmol} \, \hbox {cell}^{-1} \, \hbox {h}^{-1}\))

\(q_{\mathrm{Lac,uptake}} \) :

Uptake rate of lactate (\(\hbox {mmol} \, \hbox {cell}^{-1} \, \hbox {h}^{-1}\))

\(q_{\mathrm{Lac,uptake,max}}\) :

Maximum uptake rate of lactate (\(\hbox {mmol} \, \hbox {cell}^{-1} \, \hbox {h}^{-1}\))

\(q_{\mathrm{mAb}} \) :

Antibody formation rate (\(\hbox {mg} \, \hbox {cell}^{-1} \, \hbox {h}^{-1}\))

\(R^{2}\) :

Coefficient of determination (−)

\({U}_{{i}}\) :

Upper acceptable response (−)

V :

Volume (l)

\(X_{\mathrm{d}}\) :

Dead cell density (\(\hbox {cells} \, \hbox {ml}^{-1}\))

\(X_{\mathrm{t}} \) :

Total cell density (\(\hbox {cells} \, \hbox {ml}^{-1}\))

\(X_{\mathrm{v}} \) :

Viable cell density (\(\hbox {cells} \, \hbox {ml}^{-1}\))

\(y_{{i}} \) :

Response (−)

\(Y_{\mathrm{Amm/Gln}}\) :

Yield coefficient of ammonia formation to glutamine uptake (−)

\(Y_{\mathrm{Lac/Glc}}\) :

Yield coefficient of lactate formation to glucose uptake (−)

Amm:

Ammonia

ANOVA:

Analysis of variance

CHO:

Chinese hamster ovary

DAPI:

4,6-diamidin-2-phenylindol

DMEM/F12:

Dulbecco’s Modified Eagle Medium/Nutrient Mixture F-12

DNA:

Deoxyribonucleic acid

DoE:

Design of Experiments

FITC:

Fluorescein isothiocyanate

FSC-A:

Forward scatter area

FSC-H:

Forward scatter height

Glc:

Glucose

Gln:

Glutamine

HPLC:

High-performance liquid chromatographic

IgG:

Immunoglobulin G

Lac:

Lactate

Long R3 IGF-1:

Long arginine 3-insulin-like growth factor-1

mAb:

Antibody

max:

Maximum

min:

Minimum

mDoE:

Model-assisted Design of Experiments

mRNA:

Messenger ribonucleic acid

OFAT:

One-factor-at-time

opt:

Optimum

PBS:

Phosphate-buffered saline

QbD:

Quality by Design

RMSD:

Root-mean-squared deviation

RSM:

Response-surface model

SSC-A:

Side scatter area

UV:

Ultraviolet

References

  1. Kumar V, Bhalla A, Rathore AS (2014) Design of experiments applications in bioprocessing: concepts and approach. Biotechnol Prog 30:86–99. https://doi.org/10.1002/btpr.1821

    Article  CAS  PubMed  Google Scholar 

  2. Mandenius C-F, Graumann K, Schultz TW, Premstaller A, Olsson I-M, Petiot E, Clemens C, Welin M (2009) Quality-by-design for biotechnology-related pharmaceuticals. Biotechnol J 4:600–609. https://doi.org/10.1002/biot.200800333

    Article  CAS  PubMed  Google Scholar 

  3. US Food and Drug Administration and others (2009) Guidance for industry: Q8 (R2) pharmaceutical development. Center for Drug Evaluation and Research https://www.ich.org, Accessed 27 Dec 2018

  4. Torkashvand F, Vaziri B, Maleknia S, Heydari A, Vossoughi M, Davami F, Mahboudi F (2015) Designed amino acid feed in improvement of production and quality targets of a therapeutic monoclonal antibody. PLOS ONE 10:1–21. https://doi.org/10.1371/journal.pone.0140597

    Article  CAS  Google Scholar 

  5. Rouiller Y, Prilleux A, Vesin M-N, Stettler M, Jordan M, Broly H (2014) Modulation of mAb quality attributes using microliter scale fed-batch cultures. Biotechnol Prog 30:571–583. https://doi.org/10.1002/btpr.1921

    Article  CAS  PubMed  Google Scholar 

  6. Zhang H, Wang H, Liu M, Zhang T, Zhang j, Wang X, Xiang W (2012) Rational development of a serum-free medium and fed-batch process for a GS-CHO cell line expressing recombinant antibody. Cytotechnology 65:363–378. https://doi.org/10.1007/s10616-012-9488-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gmeiner C, Saadati A, Maresch D, Stanimira K, Frank M, Altmann F, Herwig C, Spadiut O (2015) Development of a fed-batch process for a recombinant Pichia pastoris \(\Delta \) och1 strain expressing a plant peroxidase. Microb Cell Fact 14:1. https://doi.org/10.1186/s12934-014-0183-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ramrez J, Gutierrez H, Gschaedler A (2001) Optimization of astaxanthin production by Phaffia rhodozyma through factorial design and response surface methodology. J Biotechnol 88:259–268. https://doi.org/10.1016/S0168-1656(01)00279-6

    Article  Google Scholar 

  9. Kim SH, Lee GM (2009) Development of serum-free medium supplemented with hydrolysates for the production of therapeutic antibodies in CHO cell cultures using design of experiments. Appl Microbiol Biotechnol 83:639–648. https://doi.org/10.1007/s00253-009-1903-1

    Article  CAS  PubMed  Google Scholar 

  10. Ling WLW, Bai Y, Cheng C, Padawer I, Wu C (2015) Development and manufacturability assessment of chemically-defined medium for the production of protein therapeutics in CHO cells. Biotechnol Prog 31:1163–1171. https://doi.org/10.1002/btpr.2108

    Article  CAS  PubMed  Google Scholar 

  11. Yao C-L, Liu C-H, Chu I-M, Hsieh T-B, Hwang S-M (2003) Factorial designs combined with the steepest ascent method to optimize serum-free media for ex vivo expansion of human hematopoietic progenitor cells. Enzyme Microb Technol 33:343–352. https://doi.org/10.1016/S0141-0229(03)00144-3

    Article  CAS  Google Scholar 

  12. Montgomery DC (2017) Design and analysis of experiments. Wiley, Hoboken

    Google Scholar 

  13. Nasrabadi MRN, Razavi SH (2010) Use of response surface methodology in a fed-batch process for optimization of tricarboxylic acid cycle intermediates to achieve high levels of canthaxanthin from Dietzianatronolimnaea HS-1. J Biosci Bioeng 109:361–368. https://doi.org/10.1016/j.jbiosc.2009.10.013

    Article  CAS  Google Scholar 

  14. Abt V, Barz T, Cruz-Bournazou MN, Herwig C, Kroll P, Möller J, Pörtner R, Schenkendorf R (2018) Model-based tools for optimal experiments in bioprocess engineering. Curr Opin Chem Eng 22:244–252. https://doi.org/10.1016/j.coche.2018.11.007

    Article  Google Scholar 

  15. Möller J, Pörtner R (2017) Model-based design of process strategies for cell culture bioprocesses: state of the art and new perspectives. In: Gowder SJT (ed) New insights into cell culture technology, 1st edn. Intech, London

    Google Scholar 

  16. Mandenius C-F, Brundin A (2008) Bioprocess optimization using design-of-experiments methodology. Biotechnol Prog. https://doi.org/10.1002/btpr.67

  17. Duvar S, Hecht V, Finger J, Gullans M, Ziehr H (2013) Developing an upstream process for a monoclonal antibody including medium optimization. BMC Proc https://doi.org/10.1186/1753-6561-7-S6-P34

  18. Legmann R, Schreyer HB, Combs RG, McCormick EL, Russo AP, Rodgers ST (2009) A predictive high-throughput scale-down model of monoclonal antibody production in CHO cells. Biotechnol Bioeng 104:1107–1120. https://doi.org/10.1002/bit.22474

    Article  CAS  PubMed  Google Scholar 

  19. Moran EB, McGowan ST, McGuire JM, Frankland JE, Oyebade IA, Waller W, Archer LC, Morris LO, Pandya J, Nathan SR, Smith L, Cadette ML, Michalowski JT (2000) A systematic approach to the validation of process control parameters for monoclonal antibody production in fed-batch culture of a murine myeloma. Biotechnol Bioeng 69:242–255. 10.1002/1097-0290(20000805)69:3\(<\)242::AID-BIT2\(>\)3.0.CO;2-I

  20. von Stosch M, Hamelink J-M, Oliveira R (2016) Hybrid modeling as a QbD/PAT tool in process development: an industrial E. coli case study. Bioprocess Biosyst Eng 39:773–784. https://doi.org/10.1007/s00449-016-1557-1

    Article  CAS  Google Scholar 

  21. Amanullah A, Otero JM, Mikola M, Hsu A, Zhang J, Aunins J, Schreyer HB, Hope JA, Russo AP (2010) Novel micro-bioreactor high throughput technology for cell culture process development: reproducibility and scalability assessment of fed-batch CHO cultures. Biotechnol Bioeng 106:57–67. https://doi.org/10.1002/bit.22664

    Article  CAS  PubMed  Google Scholar 

  22. Wu P, Ray N, Shuler M (1992) A Single-cell model for CHO cells. Ann NY Acad Sci 665:152–187. https://doi.org/10.1111/j.1749-6632.1992.tb42583.x

    Article  CAS  PubMed  Google Scholar 

  23. Möhler L, Flockerzi D, Sann H, Reichl U (2005) Mathematical model of influenza a virus production in large-scale microcarrier culture. Biotechnol Bioeng 90:46–58. https://doi.org/10.1002/bit.20363

    Article  CAS  PubMed  Google Scholar 

  24. López-Meza J, Araz D, Carrillo L, López-Pacheco F, Rocha M, Alvarez M (2015) Using simple models to describe the kinetics of growth, glucose consumption, and monoclonal antibody formation in naive and infliximab producer CHO cells. 68:1287–1300, https://doi.org/10.1007/s10616-015-9889-2

  25. Frahm B, Lane P, Atzert H, Munack A, Hoffmann M, Hass VC, Pörtner R (2002) Adaptive, Model-based control by the open-loop-feedback-optimal (OLFO) controller for the effective fed-batch cultivation of hybridoma cells. Biotechnol Prog 18:1095–1103. https://doi.org/10.1021/bp020035y

    Article  CAS  PubMed  Google Scholar 

  26. Caramihai M, Severin I (2013) Bioprocess modeling and control. In: Miodrag DM (ed) Biomass now—sustainable growth and use, 1st edn. Intech, London

    Google Scholar 

  27. Provost A, Bastin G (2004) Dynamic metabolic modelling under the balanced growth condition. J Process Control 14:717–728. https://doi.org/10.1016/j.jprocont.2003.12.004

    Article  CAS  Google Scholar 

  28. Kern S, Platas-Barradas O, Pörtner R, Frahm B (2016) Model-based strategy for cell culture seed train layout verified at lab scale. Cytotechnology 68:1019–1032. https://doi.org/10.1007/s10616-015-9858-9

    Article  PubMed  Google Scholar 

  29. Amribt Z, Niu H, Bogaerts P (2013) Macroscopic modelling of overflow metabolism and model based optimization of hybridoma cell fed-batch cultures. Biochem Eng J 70:196–209. https://doi.org/10.1016/j.bej.2012.11.005

    Article  CAS  Google Scholar 

  30. Carrondo MJT, Alves PM, Carinhas N, Glassey J, Hesse F, Merten O, Micheletti M, Noll T, Oliveira R, Reichl U, Staby A, Teixeira P, Weichert H, Mandenius C (2012) How can measurement, monitoring, modeling and control advance cell culture in industrial biotechnology? Biotechnol J 7:1522–1529. https://doi.org/10.1002/biot.201200226

    Article  CAS  PubMed  Google Scholar 

  31. Kontoravdi C, Samsatli NJ, Shah N (2013) Development and design of bio-pharmaceutical processes. Curr Opin Chem Eng 2:435–441. https://doi.org/10.1016/j.coche.2013.09.007

    Article  Google Scholar 

  32. Glassey J, Gernaey KV, Clemens C, Schulz TW, Oliveira R, Striedner G, Mandenius C (2011) Process analytical technology (PAT) for biopharmaceuticals. Biotechnol J 6:369–377. https://doi.org/10.1002/biot.201000356

    Article  CAS  PubMed  Google Scholar 

  33. Kroll P, Hofer A, Stelzer IV, Herwig C (2017) Workflow to set up substantial target-oriented mechanistic process models in bioprocess engineering. Process Biochem 62:24–36. https://doi.org/10.1016/j.procbio.2017.07.017

    Article  CAS  Google Scholar 

  34. Shirsat N, English N, Glennon B, Al-Rubeai M (2015) Modelling of mammalian cell cultures. In: Al-Rubeai M (ed) Animal cell culture, 1st edn. Springer, Heidelberg

    Google Scholar 

  35. Frahm B (2014) Seed train optimization for cell culture. In: Pörtner P (ed) Animal cell biotechnology: methods and protocols, 3rd edn. Humana Press, Totowa

    Google Scholar 

  36. Vander Heiden MG, Cantley LC, Thompson CB (2009) Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324:1029–1033. https://doi.org/10.1126/science.1160809

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ozturk S, Hu W-S (2005) Cell culture technology for pharmaceutical and cell-based therapies. CRC Press, Boca Raton

    Book  Google Scholar 

  38. Glacken M, Fleischaker R, Sinskey A (1986) Reduction of waste product excretion via nutrient control: possible strategies for maximizing product and cell yields on serum in cultures of mammalian cells. Biotechnol Bioeng 28:1376–1389. https://doi.org/10.1002/bit.260280912

    Article  CAS  PubMed  Google Scholar 

  39. Hassell T, Gleave S, Butler M (1991) Growth inhibition in animal cell culture—the effect of lactate and ammonia. Appl Biochem Biotechnol 30:29–41. https://doi.org/10.1007/BF02922022

    Article  CAS  PubMed  Google Scholar 

  40. Zeng A-P, Deckwer W-D, Hu W-S (1998) Determinants and rate laws of growth and death of hybridoma cells in continuous culture. Biotechnol Bioeng 57:642–654

    Article  CAS  PubMed  Google Scholar 

  41. Hayter P, Curling E, Baines A, Jenkins N, Salmon I, Strange P, Bull A (1991) Chinese hamster ovary cell growth and interferon production kinetics in stirred batch culture. Appl Microbiol Biotechnol 34:559–564. https://doi.org/10.1007/BF00167898

    Article  CAS  PubMed  Google Scholar 

  42. Min Noh S, Park JH, Sin Lim M, Won Kim J, Lee G (2016) Reduction of ammonia and lactate through the coupling of glutamine synthetase selection and downregulation of lactate dehydrogenase-A in CHO cells. Appl Microbiol Biotechnol 101:1–11. https://doi.org/10.1007/s00253-016-7876-y

    Article  CAS  Google Scholar 

  43. Zhou M, Crawford Y, Ng D, Tung J, Pynn A, Meier A, Yuk I, Vijayasankaran N, Leach K, Joly J, Snedecor B, Shen A (2011) Decreasing lactate level and increasing antibody production in Chinese Hamster Ovary cells (CHO) by reducing the expression of lactate dehydrogenase and pyruvate dehydrogenase kinases 153:27–34, https://doi.org/10.1016/j.jbiotec.2011.03.003

  44. Omasa T, Higashiyama K, Shioya S, Suga K-I (1992) Effects of lactate concentration on hybridoma culture in lactate-controlled fed-batch operation. Biotechnol Bioeng 39:556–64. https://doi.org/10.1002/bit.260390511

    Article  CAS  PubMed  Google Scholar 

  45. Möller J, Korte K, Pörtner R, Zeng A-P, Jandt U (2018) Model-based identification of cell-cycle dependent metabolism and putative autocrine effects in antibody producing CHO cell culture. Biotechnol Bioeng 115:2996–3008. https://doi.org/10.1002/bit.26828

    Article  CAS  PubMed  Google Scholar 

  46. Kurano N, Leist C, Messi F, Kurano S, Fiechter A (1990) Growth behavior of Chinese hamster ovary cells in a compact loop bioreactor. 2. Effects of medium components and waste products. J Biotechnol 15:113–28. https://doi.org/10.1016/0168-1656(90)90055-G

    Article  CAS  PubMed  Google Scholar 

  47. Miller W, R Wilke C, Blanch H (1989) The transient responses of hybridoma cells to nutrient additions in continuous culture: II. Glutamine pulse and step changes. Biotechnol Bioeng 33:487–499. https://doi.org/10.1002/bit.260330414

    Article  CAS  PubMed  Google Scholar 

  48. Nelder JA, Mead R (1965) A simplex method for function minimization. Comput J 7:308–313. https://doi.org/10.1093/comjnl/7.4.308

    Article  Google Scholar 

  49. Cameron AC, Windmeijer FA (1997) An R-squared measure of goodness of fit for some common nonlinear regression models. J Econom 77:329–342. https://doi.org/10.1016/S0304-4076(96)01818-0

    Article  Google Scholar 

  50. Ulonska S, Kroll P, Fricke J, Clemens C, Voges R, Müller MM, Herwig C (2018) Workflow for target-oriented parametrization of an enhanced mechanistic cell culture model. Biotechnol J 13:1700395. https://doi.org/10.1002/biot.201700395

    Article  CAS  Google Scholar 

  51. Candioti LV, Zan MMD, Cmara MS, Goicoechea HC (2014) Experimental design and multiple response optimization. Using the desirability function in analytical methods development. Talanta 124:123–138. https://doi.org/10.1016/j.talanta.2014.01.034

    Article  CAS  PubMed  Google Scholar 

  52. Derringer G, Suich R (1980) Simultaneous optimization of several response variables. J Qual Technol 12:214–219. https://doi.org/10.1080/00224065.1980.11980968

    Article  Google Scholar 

  53. Beckmann T, Krämer O, Klausing S, Heinrich C, Thüte T, Büntemeyer H, Hoffrogge R, Noll T (2012) Effects of high passage cultivation on CHO cells: a global analysis. Appl Microbiol Biotechnol 94:659–671. https://doi.org/10.1007/s00253-011-3806-1

    Article  CAS  PubMed  Google Scholar 

  54. Castillo AE, Fuge G, Jandt U, Zeng A-P (2015) Growth kinetics and validation of near-physiologically synchronized HEK293s cultures. Eng Life Sci 15:509–518. https://doi.org/10.1002/elsc.201400224

    Article  CAS  Google Scholar 

  55. Wippermann A, Rupp O, Brinkrolf K, Hoffrogge R, Noll T (2015) The DNA methylation landscape of Chinese hamster ovary (CHO) DP-12 cells. J Biotechnol 199:38–46. https://doi.org/10.1016/j.jbiotec.2015.02.014

    Article  CAS  PubMed  Google Scholar 

  56. Klein T, Heinzel N, Kroll P, Brunner M, Herwig C, Neutsch L (2015) Quantification of cell lysis during CHO bioprocesses: impact on cell count, growth kinetics and productivity. J Biotechnol 207:67–76. https://doi.org/10.1016/j.jbiotec.2015.04.021

    Article  CAS  PubMed  Google Scholar 

  57. Telen D, Logist F, Derlinden EV, Tack I, Impe JV (2013) Optimal experiment design for dynamic bioprocesses: a multi-objective approach. Chem Eng Sci 78:82–97. https://doi.org/10.1016/j.ces.2012.05.002

    Article  CAS  Google Scholar 

  58. Nolan RP, Lee K (2011) Dynamic model of CHO cell metabolism. Metab Eng 13:108–124. https://doi.org/10.1016/j.ymben.2010.09.003

    Article  CAS  PubMed  Google Scholar 

  59. Jincai L, Loong WC, Natarajan V, Terry H, Ashraf A (2012) Feeding lactate for CHO cell culture processes: impact on culture metabolism and performance. Biotechnol Bioeng 109(5):1173–1186. https://doi.org/10.1002/bit.24389

    Article  CAS  Google Scholar 

  60. Hartley F, Walker T, Chung V, Morten K (2018) Mechanisms driving the lactate switch in Chinese hamster ovary cells. Biotechnol Bioeng 115:1890–1903. https://doi.org/10.1002/bit.26603

    Article  CAS  PubMed  Google Scholar 

  61. Torres M, Altamirano C, Dickson AJ (2018) Process and metabolic engineering perspectives of lactate production in mammalian cell cultures. Curr Opin Chem Eng 22:184–190. https://doi.org/10.1016/j.coche.2018.10.004

    Article  Google Scholar 

  62. Gagnon M, Hiller G, Luan Y-T, Kittredge A, DeFelice J, Drapeau D (2011) High-End pH-controlled delivery of glucose effectively suppresses lactate accumulation in CHO Fed-batch cultures. Biotechnol Bioeng 108:1328–1337. https://doi.org/10.1002/bit.23072

    Article  CAS  PubMed  Google Scholar 

  63. Fu T, Zhang C, Jing Y, Jiang C, Li Z, Wang S, Ma K, Zhang D, Hou S, Dai J, Kou G, Wang H (2016) Regulation of cell growth and apoptosis through lactate dehydrogenase C over-expression in Chinese hamster ovary cells. Appl Microbiol Biotechnol 100:5007–5016. https://doi.org/10.1007/s00253-016-7348-4

    Article  CAS  PubMed  Google Scholar 

  64. Andersen DC, Goochee CF (1995) The effect of ammonia on the O-linked glycosylation of granulocyte colony-stimulating factor produced by chinese hamster ovary cells. Biotechnol Bioeng 47:96–105. https://doi.org/10.1002/bit.260470112

    Article  CAS  PubMed  Google Scholar 

  65. Ha TK, Kim Y-G, Lee GM (2015) Understanding of altered N-glycosylation-related gene expression in recombinant Chinese hamster ovary cells subjected to elevated ammonium concentration by digital mRNA counting. Biotechnol Bioeng 112:1583–1593. https://doi.org/10.1002/bit.25568

    Article  CAS  PubMed  Google Scholar 

  66. Li Z, Hong S, Yuanxing Z (2004) Fed-batch culture of hybridoma cells in serum-free medium using an optimized feeding strategy. J Chem Technol Biot 79:171–181. https://doi.org/10.1002/jctb.940

    Article  CAS  Google Scholar 

  67. Pörtner R, Schwabe J-O, Frahm B (2004) Evaluation of selected control strategies for fed-batch cultures of a hybridoma cell line. Biotechnol Appl Bioc 40:47–55. https://doi.org/10.1042/BA20030168

    Article  Google Scholar 

  68. Kildegaard HF, Fan Y, Sen JW, Larsen B, Andersen MR (2016) Glycoprofiling effects of media additives on IgG produced by CHO cells in fed-batch bioreactors. Biotechnol Bioeng 113:359–366. https://doi.org/10.1002/bit.25715

    Article  CAS  PubMed  Google Scholar 

  69. Hiller GW, Ovalle AM, Gagnon MP, Curran ML, Wang W (2017) Cell-controlled hybrid perfusion fed-batch CHO cell process provides significant productivity improvement over conventional fed-batch cultures. Biotechnol Bioeng 114:1438–1447. https://doi.org/10.1002/bit.26259

    Article  CAS  PubMed  Google Scholar 

  70. Selvarasu S, Ho YS, Chong WPK, Wong NSC, Yusufi FNK, Lee YY, Yap MGS, Lee D-Y (2012) Combined in silico modeling and metabolomics analysis to characterize fed-batch CHO cell culture. Biotechnol Bioeng 109:1415–1429. https://doi.org/10.1002/bit.24445

    Article  CAS  PubMed  Google Scholar 

  71. Lu F, Toh PC, Burnett I, Li F, Hudson T, Amanullah A, Li J (2013) Automated dynamic fed-batch process and media optimization for high productivity cell culture process development. Biotechnol Bioeng 110:191–205. https://doi.org/10.1002/bit.24602

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was partially funded by the project: Federal Ministry of Education and Research, Germany, under Grant nos. 031B0305 and 031B0577A “New mDoE-Software-Toolbox for model-based optimization of biotechnological processes”.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ralf Pörtner.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 1781 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Möller, J., Kuchemüller, K.B., Steinmetz, T. et al. Model-assisted Design of Experiments as a concept for knowledge-based bioprocess development. Bioprocess Biosyst Eng 42, 867–882 (2019). https://doi.org/10.1007/s00449-019-02089-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00449-019-02089-7

Keywords

Navigation