macroscopic models of chinese hamster ovary cell...

163
Macroscopic models of Chinese hamster ovary cell cultures ERIKA HAGROT Doctoral Thesis in Biotechnology KTH Royal Institute of Technology School of Engineering Sciences in Chemistry, Biotechnology and Health Department of Industrial Biotechnology Stockholm, Sweden 2019

Upload: others

Post on 12-Oct-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Macroscopic models ofChinese hamster ovary cell cultures

ERIKA HAGROT

Doctoral Thesis in BiotechnologyKTH Royal Institute of TechnologySchool of Engineering Sciences in Chemistry, Biotechnology and HealthDepartment of Industrial BiotechnologyStockholm, Sweden 2019

Page 2: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

TRITA-CBH-FOU-2019:45ISBN 978-91-7873-315-6

School of Engineering Sciences in Chemistry, Biotechnology and HealthKTH Royal Institute of TechnologyDepartment of Industrial Biotechnology

AlbaNova University CentreSE-106 91 Stockholm, Sweden

Academic dissertation which, with due permission from KTH Royal Institute ofTechnology, is submitted in partial fulfillment of the requirements for the degreeof Doctor of Philosophy in Biotechnology. The public review will be held onWednesday 13th November 2019 at 10:00, FB54, Roslagstullsbacken 21, Stockholm,Sweden.

© Erika Hagrot, 2019

Print: Universitetsservice US-AB, Stockholm, 2019

Page 3: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Abstract

Biopharmaceuticals treat a range of diseases, and is a growing sectorwithin the pharmaceutical industry. A majority of these complex moleculesare produced by genetically modified mammalian cells in large-scale cellcultures. Biopharmaceutical process development is costly and labor intensive,and has often been based on time-consuming empirical methods and trial-and-error. Mathematical modeling has great potential to speed up this work. Acentral question however, engaging researchers from various fields, is how totranslate these complex biological systems into feasible and useful models.

For biopharmaceutical production, macroscopic kinetic flux modeling hasbeen proposed. This model type is derived from typical data obtained in theindustry, and has been able to simulate cell growth and the uptake/secretionof important metabolites. Often, however, their scope is limited to specificculture conditions due to e.g. the lack of information on reaction kinetics,limited data sets, and simplifications to achieve calculability.

In this thesis, the macroscopic kinetic model type is the starting point, butthe goal is to capture a variety of culture conditions, as will be necessary forfuture applications in process optimization. The e↵ects of varied availabilityof amino acids in the culture medium on cell growth, uptake/secretion ofmetabolites, and product secretion were studied in cell cultures.

In Paper I, the established methodology of Metatool was tested: (i) asimplified metabolic network of approx. 30 reactions was defined; (ii) allpossible so-called elementary flux modes (EFMs) through the network wereidentified using an established mathematical algorithm; and (iii) the e↵ecton each flux was modelled by a simplified generalized kinetic equation. Alimitation was identified; the Metatool algorithm could only handle simplenetworks, and therefore several reactions had to be discarded. In this paper,a new strategy for the kinetics was developed. A pool of alternative kineticequations was created, from which a smaller set could be given higher weightas determined via data-fitting. This improved the simulations.

The identification of EFMs was further studied in papers II–IV. In Paper II,a new algorithm was developed based on the column generation optimizationtechnique, that in addition to the network also accounts for the data from oneof the parallel cultures. The method identifies a subset of the EFMs that canoptimally fit the data, even in more complex metabolic networks.

In Paper III, a kinetic model based on EFM subsets in a 100 reactionnetwork was generated, which further improved the simulations. Finally, inPaper IV, the algorithm was extended to EFM identification in a genome-scalenetwork. Despite the high complexity, small subsets of EFMs relevant to theexperimental data could be e�ciently identified.Keywords: Chinese hamster ovary; Amino acid; Metabolic network; Metabolic fluxanalysis; Kinetic modeling; Elementary flux mode; Optimization; Column generation

iii

Page 4: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Sammanfattning

Bioläkemedel används vid behandling av en rad olika sjukdomar ochutgör därför en växande sektor inom läkemedelsbranschen. Majoriteten avdessa läkemedel produceras via storskalig cellodling av genetiskt modifierademammalieceller. Processutvecklingen är dyr och arbetskrävande, och baserasvanligtvis på empirisk erfarenhet och trial-and-error. Matematiska modellerhar stor potential för att e↵ektivisera arbetet. En central fråga är dock hur manska kunna översätta ett så pass komplext biologiskt system till en genomförbaroch användbar modell.

För bioläkemedelsproduktion har s. k. makroskopisk kinetisk flödes-modellering föreslagits. Modelltypen bygger på den typ av data som tas framinom industrin och modellerna har visats kunna simulera celltillväxten, samtcellernas upptag och utsöndring av viktiga metaboliter. Dock är tillämpnings-området ofta begränsat till specifika odlingsvillkor, delvis p.g.a. kunskapsbristgällande reaktionskinetiken, begränsad tillgång till odlingsdata, samt behovetav beräkningsmässiga förenklingar.

Denna avhandling tar avstamp i makroskopisk kinetisk modellering, menhär med målet att fånga upp de mer varierade odlingsvillkor som behövs föratt kunna optimera processer. En cellinje studerades först i parallella odlingarmed varierad tillgång på aminosyror. Påverkan på tillväxt, upptag/utsöndringav metaboliter och läkemedelsproduktion registrerades.

I artikel I prövades metodiken etablerad i tidigare studier: (i) ett förenklatmetaboliskt flödesnätverk om cirka 30–40 reaktioner togs fram; (ii) samtligas.k. elementära flöden genom nätverket identifierades med en etablerad mate-matisk algoritm; (iii) påverkan på varje flöde beskrevs av en förenklad ochallmän kinetisk ekvation. Dels klarade algoritmen endast mycket förenkla-de nätverk och ett flertal reaktioner kunde därför inte tas med, dels var denkinetiska ekvationen alltför begränsad för att kunna simulera många av flödes-förändringarna i datan. Därför togs en ny strategi för kinetiken fram i artikel I.En pool av alternativa ekvationer skapades, varifrån ett mindre antal kundeges större vikt via dataanpassning. Detta förbättrade simuleringsresultaten.

Identifieringen av elementära flöden studerades sedan i artiklarna II–IV. III togs en ny algoritm fram, baserad på en optimeringsteknik kallad kolumn-generering. Algoritmen identifierar en delmängd av de elementära flödenagenom ett givet nätverk, med målet att uppnå optimal dataanpassning för enenskild odling. Detta visade sig vara e↵ektivt även för mer komplexa nätverk.I III tillämpades metoden för att simulera samtliga odlingar tillsammans i enenda modell. Den kinetiska modellen kunde nu baserades på en delmängd avflödena i ett stort nätverk om cirka 100 reaktioner, vilket förbättrade simu-leringsresultaten ytterligare. I IV, utvidgades till sist den nya algoritmen föridentifiering i en genomskalig modell. Trots den höga nivån av komplexitetkunde små delmängder av elementära flöden e↵ektivt tas fram.

iv

Page 5: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

List of papers and authors’ contributions

I Poly-pathway model, a novel approach to simulate multiple metabolic states byreaction network–based model — Application to amino acid depletion in CHOcell culture

Erika Hagrot, Hildur Æsa Oddsdóttir, Joan Gonzalez Hosta, Elling W. Jacobsen, andVéronique ChotteauPublished in Journal of Biotechnology, vol. 259, 2017, pp. 235–247DOI: 10.1016/j.jbiotec.2017.05.026

II On dynamically generating relevant elementary flux modesin a metabolic network using optimization

Hildur Æsa Oddsdóttir, Erika Hagrot, Véronique Chotteau, and Anders ForsgrenPublished in Journal of Mathematical Biology, vol. 71, 2015, pp. 903–920DOI: 10.1007/s00285-014-0844-1

III Novel column generation-based optimization approachfor poly-pathway kinetic model applied to CHO cell culture

Erika Hagrot, Hildur Æsa Oddsdóttir, Anders Forsgren, Meeri Mäkinen, and Véronique ChotteauPublished in Metabolic Engineering Communications, vol. 8, 2019, e00083DOI: 10.1016/j.mec.2018.e00083

IV Identification of experimentally relevant elementary flux mode subsetsin a genome-scale metabolic network of CHO cell metabolismusing column generation

Erika Hagrot, Hildur Æsa Oddsdóttir, Anders Forsgren, and Véronique ChotteauManuscript, 2019

The above papers will be referred to by their roman numerals. The following paper is relatedto the work, but not included in this thesis:

V Robustness analysis of elementary flux modesgenerated by column generationHildur Æsa Oddsdóttir, Erika Hagrot, Véronique Chotteau, and Anders ForsgrenPublished in Mathematical Biosciences, vol. 273, 2016, pp. 45–46DOI: 10.1016/j.mbs.2015.12.009

v

Page 6: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

The thesis project has been carried out in collaboration with the Division ofOptimization and Systems Theory at the KTH Mathematics Department. The workwas carried out by Erika Hagrot (EH), supervised by Dr. Véronique Chotteau (VC);and Hildur Æsa Oddsdóttir (HÆO), supervised by Prof. Anders Forsgren (AF),within their respective doctoral studies.

Paper I

EH planned and executed the experiments, constructed metabolic networks, de-veloped a Matlab–based framework (for handling experimental data, metabolicnetworks and kinetic modeling), performed the modeling work and wrote themanuscript. HÆO contributed in formalization of mathematical descriptions, imple-mentation of kinetic modeling, and in finalizing the manuscript.

Paper II

HÆO developed mathematical theory and novel algorithms, implemented thesealgorithms in Matlab, and wrote the manuscript. EH contributed with experimentaldata, biological descriptions and interpretation, and significantly to the style, pre-sentation, and language.

Paper III

Paper III is based on the modeling approach in Paper I and on the mathematicaltheory and novel algorithms developed in Paper II. The work was a continuation ofPaper D in the doctoral thesis of HÆO (Oddsdóttir, 2015). EH performed additionalmodel development and wrote the manuscript.

Paper IV

Paper IV is based on the mathematical theory and novel algorithms developedin Paper II. EH implemented additional features in these algorithms, applied themethod to a genome-scale network, and wrote the manuscript.

vi

Page 7: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Abbreviations and notationsAbbreviations

C. griseus Cricetulus griseusCEAA Conditionally essential amino acidCG Column generationCHO Chinese hamster ovaryE. coli Escherichia coliEAA Essential amino acidEC Extreme currentEFM Elementary flux modeEP Extreme pathwayFBA Flux balance analysisFVA Flux variability analysisGLC GlucoseGLN GlutamineHPLC High-performance liquid chromatographyIgG Immunoglobulin GKEGG Kyoto Encyclopedia of Genes and GenomesLSQ Least-squares (a technique of equation fitting)mAb Monoclonal antibodyMacro-reaction Macroscopic reactionMDH Malate dehydrogenaseME Malic enzymeMFA Metabolic flux analysisMG Minimal generatorMM Michaelis-MentenMP Master problemMVC/mL Million viable cells per milliliter, 106 viable cells/mLNEAA Non-essential amino acidPC Pyruvate carboxylasePEPck Phosphoenolpyruvate carboxykinasePPP Pentose phosphate pathwayPTMs Post-translational modificationsSP SubproblemTCA-cycle Tricarboxylic acid cycle

vii

Page 8: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Abbreviations for the amino acids

Ala A AlanineArg R ArginineAsn N AsparagineAsp D AspartateCys C CysteineGlu E GlutamateGln Q GlutamineGly G GlycineHis H HistidineIle I IsoleucineLeu L LeucineLys K LysineMet M MethioninePhe F PhenylalaninePro P ProlineSer S SerineThr T ThreonineTrp W TryptophanTyr Y TyrosineVal V Valine

viii

Page 9: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Other metabolites of the metabolic networks

3PG 3-phosphoglycerateAcCoA Acetyl-CoAAcetoAc AcetoacetateAcetoAcCoA Acetoacetyl-CoAaKad ↵-KetoadipateaKbut ↵-KetobutyrateaKG ↵-KetoglutarateArgSucc ArgininosuccinateATP Adenosine triphosphateBiomassext BiomassCho CholineChoext Choline (extracellular)Cholesterol CholesterolCit CitrateCln CitrullineCO2 Carbon dioxideCO2ext Carbon dioxide (extracellular)DHAP Dihydroxyacetone phosphateDNA Deoxyribonucleic acidE4P Erythrose-4-phosphateEthn EthanolamineEthnext Ethanolamine (extracellular)F6P Fructose-6-phosphateFADH2 Flavin adenine dinucleotideFum FumarateG3P Glyceraldehyde-3-phosphateG6P Glucose-6-phosphateGA3P Glyceraldehyde-3-phosphateGlc GlucoseGlcext Glucose (extracellular)GluySA Glutamate-�-semialdehydeGlyc3P Glycerol-3-phosphateGTP Guanosine-5’-triphosphateIMP Inosine monophosphate

ix

Page 10: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Other metabolites of the metabolic networks (continued)

Lac LactateLacext Lactate (extracellular)mAbext Monoclonal antibody (extracellular)Mal MalateLipids Membrane lipidsNADH/NAD+ Nicotinamide adenine dinucleotideNADPH/NADP+ Nicotinamide adenine dinucleotide phosphateNH4 Ammonium, NH4

+

NH4ext Ammonium (extracellular), NH4+

Orn OrnitineOrot OrotateOxa OxaloacetatePEP PhosphoenolpyruvatePhosphC phosphatidylcholinePhosphE phosphatidylethanolaminePhosphS PhosphatidylserinePropCoA Propionyl-CoAPRPP Phosphoribosyl-pyrophosphatePyr PyruvateR5P Ribose-5-PRl5P Ribulose-5-PRNA Ribonucleic acidSphm SphingomyelinSuc SuccinateSucCoA Succinyl-CoATC Total carbohydratesUrea UreaUreaext Urea (extracellular)X5P Xylulose-5-phosphate

x

Page 11: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Notations

A Stoichiometric matrixAext Stoichiometric matrix for extracellular metabolitesAextnm Stoichiometric matrix for unmeasured extracellular metabolitesAint Stoichiometric matrix for intracellular metabolitesAmac Macroscopic stoichiometric matrix (for extracellular metabolites)ai, j Stoichiometric coe�cient for the i:th metabolite and the j:th

reaction in ACtot Total cell concentration, total cell densityCv Viable cell concentration, viable cell densityCv,0 Initial viable cell concentration in an experimentc Concentrations of metabolitesc0 Initial concentrations of metabolites in an experimentci Concentration of metabolite ici,0 Initial concentration of metabolite i in an experimentE Matrix of elementary flux modesEB Matrix of a subset of the elementary flux modes in a network

(that are active)EN Matrix of a subset of the elementary flux modes in a network

(that are inactive)e⇤ An optimal solution to the SPeirrev Vector indicating if an EFM is reversible (0) or irreversible (1)el Elementary flux mode, the l:th column in Ee j,l Coe�cient for the j:th reaction and the l:th EFM in EF Matrix of kinetic functionsf j Kinetic function for the j:th reactionfl Kinetic function for the l:th EFM/macroscopic reactionfob j Objective function in flux balance analysis (FBA)G MediaJ ReactionsJrev Reversible reactionsJirrev Irreversible reactionsK Matrix with kinetic parametersKp Product inhibition constant/parameterKs Substrate saturation constant/parameterKr Metabolite inhibition constant/parameterL Pathways/elementary flux modes/Macroscopic reactions

xi

Page 12: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Notations (continued)

Mall All metabolitesMext Extracellular metabolitesMextnm Unmeasured extracellular metabolitesMint Intracellular metabolitesP Matrix of pathways (or extreme rays)P Product/Product concentrationpl Pathway, the l:th column in Pp j,l Coe�cient for the j:th reaction and the l:th pathway in Pqext Cell-specific rates for measured extracellular metabolitesqextnm Cell-specific rates for unmeasured extracellular metabolitesqsim Simulated cell-specific ratesqext,i Cell-specific uptake/secretion rate for metabolite iqextnm,i Cell-specific uptake/secretion rate for unmeasured metabolite iqmAb Cell-specific mAb production rateS Substrate/Substrate concentrationt Time pointt0 Initial time point in an experimentv Flux ratesvirrev Vector indicating if a reaction is reversible (0) or irreversible (1)v j Cell-specific flux rate for the j:th reactionvmax, j Maximum cell-specific flux rate for the j:th reactionw Macroscopic flux ratesw⇤ An optimal solution to the EFMs-based MFA problemwB An solution to the EFMs-based MFA problem with a subset of

the EFMsw⇤B An optimal solution to the EFMs-based MFA problem with a

subset of the EFMswmax Maximum macroscopic flux rateswl Cell-specific macroscopic flux rate for the l:th macroscopic reac-

tionwmax,l Maximum cell-specific macroscopic flux rate for the l:th macro-

scopic reactionµ Cell-specific growth rate

xii

Page 13: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

List of Figures

2.1 The lineage of popular CHO cell lines . . . . . . . . . . . . . . . . . . . . . . 112.2 Overview of the cell metabolism . . . . . . . . . . . . . . . . . . . . . . . . 202.3 The connection between glutaminolysis and glycolysis in CHO cells . . . . . . 27

3.1 A macroscopic/black-box model of a cell population . . . . . . . . . . . . . 353.2 Comparison of the network sizes in the present work . . . . . . . . . . . . . 483.3 Conceptual illustration of the flux space . . . . . . . . . . . . . . . . . . . . 503.4 Example of a simple pathway linking glucose to lactate . . . . . . . . . . . . 543.5 Graphical illustration of the extreme rays . . . . . . . . . . . . . . . . . . . 563.6 A comparison of EPs versus EFMs . . . . . . . . . . . . . . . . . . . . . . 603.7 Plot of a Michaelis-Menten–type equation . . . . . . . . . . . . . . . . . . . 653.8 Plot of saturation and inhibition for di↵erent parameter values . . . . . . . . 693.9 Workflow for a derivation of a macroscopic kinetic model . . . . . . . . . . 73

4.1 Workflow of a parallel cell culture experiment . . . . . . . . . . . . . . . . . 764.2 Pseudo-perfusion protocol with example of culture data . . . . . . . . . . . 78

5.1 The three principles for the metabolic model . . . . . . . . . . . . . . . . . 845.2 The one-model approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . 855.3 The workflow toward proof-of-concept in Paper I . . . . . . . . . . . . . . . . 875.4 The metabolic network map from Paper I . . . . . . . . . . . . . . . . . . . 895.5 A consideration of the kinetics at balanced growth . . . . . . . . . . . . . . . 915.6 The flexible kinetics strategy . . . . . . . . . . . . . . . . . . . . . . . . . . 92

6.1 Active/inactive EFM subsets and evaluation of a candidate EFM . . . . . . . 986.2 Workflow of the column generation-based algorithm . . . . . . . . . . . . . 1006.3 EFM enumeration versus EFM subset identification . . . . . . . . . . . . . . 102

7.1 The metabolic network map from Paper III . . . . . . . . . . . . . . . . . . 1067.2 Integration of column generation into the modeling framework . . . . . . . . . 1077.3 Weighting factors to improve the fit for unusual behavior . . . . . . . . . . . . 1117.4 The flexible kinetics strategy, now with model reduction . . . . . . . . . . . 112

xiii

Page 14: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7.5 Overview of the cross-validation and model selection in Paper III. . . . . . . 113

xiv

Page 15: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

List of Tables

2.1 The reactions of the glycolysis pathway . . . . . . . . . . . . . . . . . . . . . 212.2 The reactions of the pentose phosphate pathway . . . . . . . . . . . . . . . . 222.3 The reactions of the TCA-cycle . . . . . . . . . . . . . . . . . . . . . . . . 232.4 The reactions of the essential amino acid metabolism . . . . . . . . . . . . . 252.5 The reactions of the non-essential amino acid metabolism . . . . . . . . . . 262.6 The anaplerotic reactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . 282.7 Possible amino acid syntheses . . . . . . . . . . . . . . . . . . . . . . . . . 292.8 The reactions of the urea cycle . . . . . . . . . . . . . . . . . . . . . . . . . 302.9 Patterns of amino acid uptake/secretion in CHO cells . . . . . . . . . . . . . . 31

3.1 Three types of idealizations that enable the modeling of complex model targets 373.2 Five guiding ideals that govern the model building process . . . . . . . . . . . 373.3 A framework for the classification of cell population models . . . . . . . . . 403.4 An overview of the metabolic networks in the present work . . . . . . . . . . . 473.5 Tasks of EFM analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 583.6 Publications focused on kinetic modeling of cell cultures . . . . . . . . . . . 64

4.1 The selected amino acids to vary in the experiment . . . . . . . . . . . . . . 794.2 The selected concentration levels for the experiment . . . . . . . . . . . . . 80

6.1 Important contributions to the development of column generation . . . . . . . 97

7.1 Kinetic model development in Paper III . . . . . . . . . . . . . . . . . . . . 110

8.1 Extracting information from the EFMs and the macroscopic reactions . . . . . 121

xv

Page 16: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work
Page 17: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Contents

Abstracts iii

List of papers and authors’ contributions v

Abbreviations and notations vii

List of Figures xiii

List of Tables xv

1 Introduction 11.1 Production of biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . 11.2 Challenges in process development . . . . . . . . . . . . . . . . . . . . 21.3 Mathematical modeling . . . . . . . . . . . . . . . . . . . . . . . . . . 31.4 Modeling approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . 31.5 Aim of the present thesis . . . . . . . . . . . . . . . . . . . . . . . . . . 51.6 Thesis organization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6

2 Biopharmaceutical production 72.1 Biopharmaceutical products and market . . . . . . . . . . . . . . . . . . . 72.2 Production and process development . . . . . . . . . . . . . . . . . . . 82.3 The metabolism of the CHO cell lines . . . . . . . . . . . . . . . . . . . 182.4 Future strategies for process optimization with metabolic models . . . . 32

3 Mathematical modeling of cells 353.1 Idealization and guiding ideals in the model construction process . . . . 363.2 Model classifications . . . . . . . . . . . . . . . . . . . . . . . . . . . . 393.3 Introduction to metabolic reaction networks . . . . . . . . . . . . . . . . 423.4 Introduction to metabolic flux analysis (MFA) . . . . . . . . . . . . . . 463.5 Introduction to pathway analysis . . . . . . . . . . . . . . . . . . . . . . 533.6 Kinetic modeling of cell populations . . . . . . . . . . . . . . . . . . . 623.7 Derivation of a macroscopic kinetic model . . . . . . . . . . . . . . . . 72

4 Experimental data: Influence of varied amino acid availability in CHO cellcultures 754.1 Aim and overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 754.2 Experimental plan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 774.3 Experimental data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80

xvii

Page 18: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5 Contribution of Paper I: Development of an initial methodology 835.1 Proposed concept and workflow . . . . . . . . . . . . . . . . . . . . . . 835.2 Application and further development . . . . . . . . . . . . . . . . . . . 865.3 Summary of the contribution . . . . . . . . . . . . . . . . . . . . . . . . 93

6 Contribution of Paper II: A column generation-based method for pathwayidentification in complex metabolic networks 956.1 Introduction to mathematical optimization . . . . . . . . . . . . . . . . 956.2 Column generation applied to EFM identification . . . . . . . . . . . . . . 976.3 Summary of the contribution . . . . . . . . . . . . . . . . . . . . . . . . 104

7 Contribution of Paper III: Integration of the column-generation-based path-way identification into the poly-pathway methodology 1057.1 Network and identification of EFMs for the model . . . . . . . . . . . . 1057.2 Kinetic model development and evaluation . . . . . . . . . . . . . . . . 1097.3 Summary of the contribution . . . . . . . . . . . . . . . . . . . . . . . . 114

8 Contribution of Paper IV: Pathway identification in a genome-scale CHOmetabolic network using the column generation-based approach 1178.1 The genome-scale network model . . . . . . . . . . . . . . . . . . . . . . 1178.2 Pathway analysis at genome-scale . . . . . . . . . . . . . . . . . . . . . 1198.3 Summary of the contribution . . . . . . . . . . . . . . . . . . . . . . . . 120

9 Conclusions and future perspectives 123

References 127

Acknowledgments 145

xviii

Page 19: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Chapter 1

Introduction

1.1 Production of biopharmaceuticalsOver the last decades, novel biotechnological approaches have enabled the selectivegeneration of therapeutic proteins in living cells. Nowadays, high-quality complexbiopharmaceutical products are produced in large-scale cell-based bioprocesses.These biopharmaceutical products o↵er treatments for a range of potentially seriousand/or chronic conditions, and are of major importance to healthcare systemsglobally (Ecker et al., 2015). Biopharmaceuticals constitutes a fast-growing sectorin the pharmaceutical industry, generating considerable profits each year (Walsh,2014; Ecker et al., 2015; Faustino Jozala et al., 2016; Moorkens et al., 2017;Soelkner, 2017). In 2016, total global sales amounted to $228 billions (Moorkenset al., 2017), and are projected to reach $390 billions by 2020 as reported by theIMS Health market research firm (Soelkner, 2017).

Mammalian cell lines have become the preferred hosts for many biopharmaceu-tical products, in particular for the more complex ones requiring post-translationalmodifications (PTMs) (Butler and Spearman, 2014; Jedrzejewski et al., 2014; Du-mont et al., 2016). Chinese hamster ovary (CHO) cell lines derived from the ovarytissue of a Chinese hamster (Puck, Theodore et al., 1958; Wurm, 2013) (Cricetulusgriseus) are the most widely used (Wurm and Hacker, 2011; Kim et al., 2012; Walsh,2014). Products based on monoclonal antibodies (mAbs) (Leavy, 2010; Ecker et al.,2015; Pierpont et al., 2018) dominate the market, and a majority are produced inCHO cell lines (Walsh, 2014).

1

Page 20: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

1. Introduction

1.2 Challenges in process developmentUpstream process development begins with gene cloning, choice of cell line, andsubsequent cell line development (Zhang, 2010; Kim et al., 2012; Wurm, 2013;Dahodwala et al., 2012). After follows process design: the choice of bioreactor typeand cultivation mode (Xie and Wang, 1994a; Xie and Zhou, 2006; Kompala andSadettin, 2006; Henry et al., 2008), the development of suitable media (Burgenerand Butler, 2006), and the determination of various process parameters to achievesatisfactory growth rates, productivity and product quality (Li et al., 2010; Pörtneret al., 2017; Huang et al., 2017). First-to-market and product cost-e↵ectivenessare factors driving process development (Li et al., 2010; Bhambure et al., 2011).Meanwhile, the work is often complex, time-consuming, and costly (Li et al., 2010).

CHO cells and other mammalian cell lines need to be cultured in complex media,frequently of proprietary composition (Zhang, 2010) and consisting of up to 50–60di↵erent components. Genetic and phenotypic diversity can be found among thevarious cell lines in use today (Wurm and Hacker, 2011; Wurm, 2013; Kaas et al.,2014). As the chosen cell line is developed to express a novel product, additionalchanges in genome and behavior occur, leading to specific nutritional requirements(Carrillo-Cocom et al., 2015). Consequently, medium development constitutes animportant step in the upstream process development, especially since changes inan original candidate medium composition can have dramatic e↵ects on growth,product yield and/or quality (Zhang, 2010).

Mammalian cells lines are characterized by an ine�cient yet flexible metabolism(Gódia and Cairó, 2006; Sidorenko et al., 2008; Altamirano et al., 2013; Feichtingeret al., 2016). The impact of the culture media on the growth and cell metabolism incultivation can be evaluated based on the exchange of selected metabolites betweenthe cells and the medium (Kaas et al., 2014). Glucose and glutamine are the mainsubstrates and are rapidly consumed. Lactate and ammonium are rapidly secretedby-products, and potentially toxic at high concentrations. Amino acids, studiedin the present thesis, are necessary for cell growth and product formation. All 20natural amino acids are typically supplied, and their availability and concentrationscan influence growth and metabolism (Chen and Harcum, 2005). The uptake,secretion, and potential depletion of amino acids and other metabolites (Gódia andCairó, 2006; Wahrheit et al., 2014a) provide clues on how to improve media andfeeds of a particular process (Xing et al., 2011; Sellick et al., 2011).

2

Page 21: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

1.3. Mathematical modeling

1.3 Mathematical modelingWhile process development strives to optimize multiple factors, this can be di�cultto achieve through experiments and intuitive reasoning alone. To speed up the workand cut costs, screening experiments in simple small-scale culture systems and lab-scale instrumented bioreactors serve as simplified models before scale-up (Lara et al.,2017). Unfortunately, there is often a lack of understanding for why certain changesin the process conditions lead to improvements (Lewis et al., 2016; Huang et al.,2017). Mathematical modeling has the potential to accelerate this work (Zeng andBi, 2006; Bailey, 1998). Models can provide frameworks for the interpretation ofdata, enable in silico predictive simulations decreasing experimental workload, andaid the identification of optimal process conditions through optimization algorithms(Zeng and Bi, 2006). A meaningful research question, which is also the main themein this thesis, is therefore how to generate models of cell cultures and specificallythe type of models that can: (i) deepen our knowledge about cell-based processes;and (ii) ultimately function as computational tools for process development andoptimization (Bhambure et al., 2011; Almquist et al., 2014).

1.4 Modeling approachesFor cell cultures, metabolic and mechanistically-based models are preferred, inparticular for predictions outside an experimental range. Models based on generalmathematical relationships, e.g. polynomial functions, are limited in this aspect.Metabolic and mechanistically-based models incorporating biological propertiesinto the model structure are better suited for understanding cell behavior, and forprediction and optimization tasks.

Metabolic flux analysis (MFA) is a method that can be used to estimate andsimulate fluxes over metabolic reaction networks, and has been applied to modeldi↵erent organisms (Niklas et al., 2010; González-Martínez et al., 2014; Lopes andRocha, 2017), including mammalian cell lines (Xie and Wang, 1996a,b; Bonariuset al., 1997; Martens, 2007; Sidorenko et al., 2008; Quek et al., 2010; Zamoranoet al., 2010; Goudar et al., 2007; Nicolae et al., 2014; Huang et al., 2017). Here,a distinction can be made between steady-state metabolic models versus kineticor dynamic metabolic models (Steuer and Junker, 2009; Ben Yahia et al., 2015;Almquist et al., 2014). Models describing single steady-states can be achieved usingbasic MFA, potentially in combination with isotope tracer experiments (Bonariuset al., 1998), or flux balance analysis (FBA) (Orth et al., 2010; Santos et al., 2011;

3

Page 22: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

1. Introduction

Martínez et al., 2013; Huang et al., 2017). FBA relies on the optimization of abiologically relevant objective function (Schuetz et al., 2007; Orman et al., 2011),and is frequently applied to simulate the steady-state metabolism of microorganismsunder the assumption of maximized growth (Feist and Palsson, 2010; Huang et al.,2017). Steady-state modeling, including FBA, has been applied to mammaliancells and cell lines in several studies (Xie and Wang, 1996a,b; Sheikh et al., 2005;Sidorenko et al., 2008; Quek et al., 2010), including CHO cells (Altamirano et al.,2006; Zamorano et al., 2010; Goudar et al., 2010; Xing et al., 2011; Martínezet al., 2013). For FBA studies of microorganisms, large and complex genome-scale consensus models derived from the organism’s genome have been employedfor many years (Edwards and Palsson, 2000; Orth et al., 2011; Feist et al., 2007;Hädicke and Klamt, 2017). These types of genome-scale models have only recentlybecome available for CHO cells (Wurm and Hacker, 2011; Xu et al., 2011; Kaaset al., 2014; Hefzi et al., 2016; Rejc et al., 2017; Huang et al., 2017).

Steady-state models provide snapshots of cell metabolism under specified condi-tions (Huang et al., 2017). If the conditions change, the metabolism may change inresponse. Metabolic models incorporating kinetics have the ability to describe suchphenomena, and support dynamic simulations and predictions, possibly outside theexperimental range. Meanwhile, the generation of realistic kinetic models presents amajor challenge due to the lack of kinetic information for most organisms (Almquistet al., 2014). Previous studies have demonstrated the simulation of dynamic cellbehavior in cell cultures using simplified kinetic models (Provost et al., 2005; Gaoet al., 2007; Zamorano Riveros, 2012; Zamorano et al., 2013). These kinetic modelsare often limited in their scope, e.g. in the number of inputs and outputs, the sizeand flexibility of the underlying metabolic network (Jungers et al., 2011; Zamoranoet al., 2013), and/or the experimental range covered. While progress has been made(Jungers et al., 2011; Zamorano et al., 2013), many aspects remain to be studied.Attempts to build large-scale kinetic models of E. coli have been made (Khodayariet al., 2014; Khodayari and Maranas, 2016; Huang et al., 2017), yet large-scalekinetic models of CHO cells still remain poorly developed (Huang et al., 2017).

Several of the proposed kinetic cell culture models are macroscopic kineticmodels (Provost et al., 2005; Gao et al., 2007; Zamorano Riveros, 2012; Zamoranoet al., 2013; Ben Yahia et al., 2015), and this is the type of model studied in thepresent thesis. Main features are: the simplification of the metabolism into multiplepathways running through the metabolic network that links observable extracellularsubstrates to products; and a description of pathway kinetics based on simplifiedkinetic equations. This approach appears to be suitable for industrial applications,in which extracellular substrates and products are routinely monitored.

4

Page 23: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

1.5. Aim of the present thesis

1.5 Aim of the present thesisThis thesis is dedicated to mathematical modeling of cell cultures, and to thequestion of how to generate such models. The aim is stated as follows:

To develop a methodology for generating one single kinetic macroscopic model,representing mAb-producing CHO cells in pseudo-perfusion cultures, with theability to simulate diverse metabolic behavior triggered by variations in the initialavailability of single amino acids in the cultivation medium.

The specific CHO cell line, the type of bioreactor, medium and cultivation modetargeted are further specified as follows:

• The cell line is CHO-K1SV (provided by Selexis, Switzerland), which pro-duces Immunoglobulin G (IgG)—this is a common mAb with applications ine.g. cancer treatment (Kato, 2016; Pierpont et al., 2018).

• Cultures are carried out in TubeSpin bioreactors (Strnad et al., 2010). This isa simple culture system suitable for screening experiments, and which canserve as simplified scale-down models of the instrumented bioreactors usedin large-scale manufacturing (Lara et al., 2017).

• The cultivation medium (provided by Irvine Scientific, CA, USA) is chemically-defined, protein-free, and has the ability to support CHO-K1SV cell growthand IgG synthesis.

• As specified in the aim above, cell cultures were carried out in pseudo-perfusion cultivation mode. This is a simplified version of a continuouscultivation mode referred to as perfusion (Kompala and Sadettin, 2006).Pseudo-perfusion can be applied to study cell cultures under relatively stableculture conditions, while using simple equipment.

The main application considered in this thesis is medium development, withfocus on the amino acids in the medium. Furthermore, the aim emphasizes that onesingle model is to capture diverse metabolic behavior. This requisite stems froma more general goal of cell culture modeling: the generation of models that are,in practice, able to predict and optimize cell culture processes, e.g. to optimize amedium composition. While this goes beyond the scope of this thesis, it is assumedthat such optimization requires that diverse metabolic behavior can be simulatedand predicted in one single kinetic model.

5

Page 24: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

1. Introduction

1.6 Thesis organizationThe modeling of complex biological systems presents a major challenge; it must berealized through concerted e↵ort and engages researchers from di↵erent fields. Inthe present case, a project was carried out in a collaboration between researchersfrom biotechnology and from applied mathematics at KTH. The thesis is writtenfrom the viewpoint of a biotechnologist with industrial applications in mind, towhom the role of the mathematics is to provide tools for solving biotechnologicalproblems that arise. After this introduction, the remaining eight chapters of thethesis are organized as follows. Chapter 2 outlines biopharmaceutical productionwith focus on upstream process development and cell metabolism. Chapter 3introduces general concepts and methods for the modeling of cells, and derives atype of model proposed in the literature, and on which the present work was based.The generation of experimental data for the purpose of method development aredescribed in Chapter 4. The development of methodology and generation of modelsare covered in Chapters 5, 6, 7, and 8, each corresponding to one of the appendedpapers. Lastly, Chapter 9 gives the conclusions, as well as prospects of how researchcould be pursued in the future.

6

Page 25: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Chapter 2

Biopharmaceutical production

2.1 Biopharmaceutical products and market

2.1.1 Biopharmaceutical products

Biological molecules play important roles in the human body (Faustino Jozalaet al., 2016). Biopharmaceuticals based on such molecules provide highly specifictreatments and diagnostics for various diseases. Major product classes includehormones, growth factors, interferons, vaccines, mAbs, enzymes, fusion proteins,gene therapy and nucleic acids. The IgG produced by the CHO-K1SV cell line inthis thesis belongs to the mAbs class. In terms of best-selling products, the mAb-based products dominate the market (Walsh, 2014); in 2013, four mAb productswere found among the top five best-selling biopharmaceuticals within the EU andUS. All four were produced in mammalian cell lines; two indicated for treatmentof Rheumatoid arthritis, and the other two for Chron’s disease and Non-Hodgkin’slymphoma, respectively (Walsh, 2014).

2.1.2 mAb-based products

Antibodies are proteins produced naturally by cells of the immune system whosefunction is to recognize and attach to specific targets found in proteins. mAb-basedbiopharmaceuticals provide means to activate, inhibit or block proteins involved indisease mechanisms, and can thus be used for diagnostic and treatment purposes.

mAbs are highly-specific and generally well-tolerated with lower risk of safetyissues compared to many other types of biopharmaceuticals (Ecker et al., 2015).This has led to the widespread usage of mAb products in the treatment of numerous

7

Page 26: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

conditions, including e.g. cancer or autoimmune diseases. Depending on thecondition, patient populations can range from a few thousands (e.g. rare bloodor auto-inflammatory diseases) to hundreds of thousands (e.g. some cancers andmultiple sclerosis) to millions (e.g. asthma and rheumatoid arthritis) (Ecker et al.,2015).

2.1.3 Future outlook

Biopharmaceuticals is a growing market and the number of products are expectedto increase (Faustino Jozala et al., 2016). mAb products have been predicted1 toreach world-wide sales of nearly $125 billions by 2020 (Ecker et al., 2015). Severalfactors drive this development, e.g. an increasing and aging world population, novelmAb technologies (Tiller and Tessier, 2015; Ecker et al., 2015), and non-inventorproduct versions (biosimilars) associated with lower research and developmentcosts. As patent protection and exclusivity rights expire (Moorkens et al., 2017),a↵ordable mAb biosimilars are expected to open up cost-sensitive markets withlarge populations in e.g. India, China, and Russia (Ecker et al., 2015).

2.2 Production and process development

2.2.1 Overview of process

The general manufacturing process for biopharmaceuticals can be divided intoupstream and downstream processing (Faustino Jozala et al., 2016; Pörtner et al.,2017). Steps for the upstream process development include the selection of a cellline, the development of that cell line to produce the biopharmaceutical product,the selection of suitable culture media, bioreactor and cultivation mode, and thedefinition of process parameters such as pH and temperature. It includes strategiesfor the scale-up of cell bank/stock cultures to seed the large-scale bioreactor (seedtrain), for controlling pH and oxygen supply, as well as for ensuring sterile condi-tions throughout all operations. The steps of downstream processing involve theharvest of the product from the culture, extraction and purification via e.g. centrifu-gation, filtering, chromatography techniques, and the formulation, quality controland packaging of the final biopharmaceutical product. The modeling approachesdeveloped in this thesis target upstream process development, and the selection ofculture media in particular.

1A prediction made in 2015.

8

Page 27: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.2. Production and process development

2.2.2 Host selection

Potential hosts include microbial cells (bacterial or yeast), fungi, animal cells(insect, mammalian, or avian), and plant cells (Dumont et al., 2016; Faustino Jozalaet al., 2016; Bertolini et al., 2016). Transgenic animals (Bertolini et al., 2016)and plants are other options. The selection depends largely on the ability of theorganism to perform the necessary PTMs of the recombinant protein. This involvesthe proper assembly and folding of the protein, and in particular the attachment ofsuitable glycan structures. The latter is referred to as glycosylation, and the resultingstructure as the glycosylation profile.

PTMs are critical for biopharmaceutical proteins as they a↵ect their structure,clinical function and e�cacy (Butler and Spearman, 2014). Additionally, inappro-priate PTMs can seriously compromise the safety of the product, as it may cause aresponse in the immune system of humans (Dumont et al., 2016). The glycosylationprofiles of biopharmaceutical glycoproteins are of particular importance, as theseare species-specific and need to be identical or at least similar to those found inhuman proteins.

Mammalian cell lines are frequently chosen for the production of biopharma-ceutical products. Between 2010 and 2014, 212 biopharmaceuticals were approvedfor EU and US markets and of those 60 % were produced in mammalian systems(Walsh, 2014). Mammalian cells have the inherent ability to perform PTMs, andto achieve human-like glycosylation profiles. Examples of mammalian cell linesin biopharmaceutical production include CHO, murine myeloma (NS0 and Sp2/0),and baby hamster kidney (BHK) (Zhang, 2010). Human cell lines are also in use orunder development (Dumont et al., 2016), e.g. the human embryonic kidney 293(HEK-293) and fibrosarcoma HT-1080.

Compared to the microbial cells, mammalian cells have been associated withtechnological limitations, e.g. slow growth, an ine�cient metabolism, complexmedium requirements and low productivity (Altamirano et al., 2013). Over time,product yields have increased by several orders of magnitude thanks to gene am-plification systems, and improvements in the process design (Jayapal et al., 2007;Wurm et al., 2008; Rader and Langer, 2015; Langer, 2018). Fast growing microbialcells with simple nutritional requirements (as e.g. E. coli, Saccharomyces cere-visiae or Pichia Pastoris) may still represent more cost-e↵ective options (Dumontet al., 2016; Faustino Jozala et al., 2016). Unfortunately, due to lack of specificenzymes, intracellular compartmentalization, and chaperon enzymes involved inprotein folding, bacterial cells cannot always achieve the PTMs required (Waegemanand Soetaert, 2011; Dumont et al., 2016). Yeast cells tend to attach mannose-rich

9

Page 28: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

glycans to proteins, which can impact the product e�cacy and safety (Dumont et al.,2016). Insect and plant cells produce complex PTMs, but the resulting glycosylationprofiles achieved by these organisms are not human-like.

2.2.3 CHO cell lines

CHO cell lines have become standard in research and industry (Jayapal et al., 2007),and are the most popular hosts in biopharmaceutical production; about one third ofthe 212 biopharmaceuticals approved between 2010 and 2014 were produced byCHO cells (Walsh, 2014).

The original CHO cell line was derived in experiments carried out in the 1950s.The aim was to find new techniques for the long-term culture of human and animalcells for applications in genetic research (Puck, Theodore et al., 1958). Tissues of theChinese hamster could be successfully grown, and reported as "particularly hardyand reliable". Cells obtained from Chinese hamster ovary tissue could be culturedat maintained growth rate for several months in a row, appearing to have undergonespontaneous immortalization (Puck, Theodore et al., 1958; Wurm, 2013). Duringthe 1960s and 1970s, CHO cells were extensively used for studies in cell genetics(Wurm, 2013). In 1986, human tissue plasminogen activator (tPA) produced inCHO cells became the first biopharmaceutical produced in mammalian cells to beregulatory approved (Zhang, 2010; Altamirano et al., 2013).

The popularity of CHO cell lines in biopharmaceutical production can be ex-plained by several factors. It is easy to transfer foreign DNA into their genome(Wurm and Hacker, 2011). They possess the capacity for e�cient PTMs, resultingin glycoproteins compatible and bioactive in humans (Kim et al., 2012). Whensuitable culture conditions are applied, this leads to rapid cell growth, at which thecells typically double in number once a day (Wurm, 2013). CHO cells can be easilyadapted for suspension culture, to regulatory approved media, and to large-scaleprocesses (Kim et al., 2012). Low productivity typical for mammalian cell lines canbe overcome by powerful gene amplification systems available for CHO cell lines(Kim et al., 2012). Finally, the long track-record as safe hosts for biopharmaceuticalproduction (Wurm and Hacker, 2011) makes it easier to obtain approval from regu-latory agencies, promoting future use of CHO cell lines as production hosts (Kimet al., 2012).

The lineage of popular CHO cell lines is illustrated in Figure 2.1. The CHO-K1SV cell line used in the experimental work of this thesis originates from CHO-K1,which was derived from the original CHO cell line in the late 1960s. As the genomesof immortalized cells are dynamic (Wurm and Hacker, 2011), di↵erent CHO cell

10

Page 29: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.2. Production and process development

Figure 2.1: The lineage of CHO cell lines illustrating the historic development of importantand widely applied CHO cell lines. EMS: ethyl methanesulfonate. Lonza and Fisher arecompanies. ATCC: American Type Culture Collection. ECACC: European Collection ofAuthenticated Cell Cultures. The cell line used in the experimental work of this thesis ismarked in grey. This figure is based on an illustration by Xu et al. (2017).

2. Biopharmaceutical production

Figure 2.1: CHO cell lines lineage illustrating the historic development of important andwidely applied CHO cell lines. This figure was adapted from the cell lineage of CHOcells illustrated by Xu et al. (2017). EMS: ethyl methanesulfonate. Lonza and Fisher arecompanies. ATCC: American Type Culture Collection. ECACC: European Collection ofAuthenticated Cell Cultures. The cell line used in the experimental work of this thesis ismarked in grey.

EMS exposure

Gamma rays

CHO-ori(Puck, 1957)

CHO variant(Tobey, 1962)

CHO-Toronto/CHO Pro-5(Thompson, 1973)

CHO-S(Tilkins, 1991)

CHO Pro3-DHFR+ (Flinto↵, 1976)

CHO-TMTXRIIIDHFR mutant

CHO-DG44 (DHFR-)(Urlaub & Chasin, 1983)

cGMP CHO-DG44(Fisher)

CHO-K1(Kao & Puck, 1968)

CHO-K1(ATCC) CHO-K1SV

(Lonza)

CHO-K1SV(Selexis)

CHO-K1SV GS-KO(Lonza)

CHO DXB11/DUKX(Urlaub & Chasin, 1980)

CHO/dhrf-

CHO-K1(ECACC)

these complex biological molecules in turn requires more simple building blocks,e.g. amino acids and other metabolites taken up from the medium or producedintracellularly from other components.

Growth is quantified by the cell-specific growth rate µ, which can be calcu-lated from an initial and final viable cell density measurement at two consecutivetime points:

µ =ln( Cv

Cv,0)

t � t0, (2.1)

where Cv,0 and Cv are the numbers of viable cells at the two consecutive time pointst0 and t. The viability indicates the balance between viable and dead cells in the

12

lines have become genetically and phenotypically diverse (Wurm, 2013; Feichtingeret al., 2016). Environmental changes promote such genetic drift, as cells with certainproperties are favored over more sensitive cells (Wurm, 2013), e.g. shifting fromadherent to suspension culture, changing the medium composition, or subjectingcells to harsh conditions in large-scale bioreactor cultures. Di↵erent chromosomalnumbers and rearrangements (Wurm, 2013; Kaas et al., 2014) can be found betweenC. griseus and di↵erent CHO cell lines (Cao et al., 2012), and even between cellsin the same cell population (Wurm and Hacker, 2011; Vclear et al., 2018). Acomparison of the genomes of several CHO cell lines and C. griseus identifiedmillions of point mutations and other genetic di↵erences that have occurred overtime (Lewis et al., 2013; Kaas et al., 2014).

11

Page 30: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

2.2.4 Cell line development

After the cell line has been chosen, the subsequent cell line development involvessteps of transfection, selection and amplification (Kim et al., 2012; Wurm, 2013;Zhang, 2010). The end result is number of clones that produce the biopharmaceuti-cal, but are genetically di↵erent to the original cell line and to each other. Theseclones can respond di↵erently to changes in the media and other culture conditions.Thus, candidate clones may need to be tested against established production plat-forms, and additional process development must often be carried out (Kim et al.,2012; Dahodwala et al., 2012).

2.2.5 Development of the cultivation process

The ultimate goal for upstream process development is to define a process thatachieves high density and viability of the cells, accompanied by a high cell-specificproductivity and final amount of the recombinant product (Huang et al., 2017; Liet al., 2010). The process must also meet requirements imposed by regulatoryagencies in terms of process reproducibility, product quality (Huang et al., 2017)and safety. Important factors that can be changed to influence process performanceinclude: changes in the media composition, temperature, and pH; the choice ofbioreactor type and cultivation mode; and the strategies chosen for feed and feedrates, oxygen (O2) and carbon dioxide (CO2) supply, and agitation.

The cell growth is one of the most important cultivation variables and is observedexperimentally as an increase in the number of cells per volume with time. Theunderlying process involves cell growth and division, which requires the synthesisof nucleotides (DNA and RNA), protein and cell membrane. The synthesis ofthese complex biological molecules in turn requires more simple building blocks,e.g. amino acids and other metabolites taken up from the medium or producedintracellularly from other components.

Growth is quantified by the cell-specific growth rate µ, which can be calcu-lated from an initial and final viable cell density measurement at two consecutivetime points:

µ =ln( Cv

Cv,0)

t � t0, (2.1)

where Cv,0 and Cv are the numbers of viable cells at the two consecutive time pointst0 and t. The viability indicates the balance between viable and dead cells in theculture, and is defined as the percentage of viable cells relative to the total number

12

Page 31: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.2. Production and process development

of cells in the culture at a specific time point.

Viability% = 100 ⇤ Cv

Ctot(2.2)

To evaluate the productivity, the cell-specific product secretion rate betweentwo consecutive time points can be calculated as follows:

qmAb = µ ·cmAb � cmAb,0

Cv �Cv,0, (2.3)

where cmAb,0 and cmAb are the concentration of mAb at two consecutive time pointst0 and t. The amount of product depends both on the cell-specific product secretionrate and the number of viable and producing cells. Even if qmAb is stable, animpaired cell growth and low viable cell density reduce the amount of product thatcan be obtained.

2.2.6 Culture conditions

As first reported by Puck, Theodore et al. (1958), and in contrast to e.g. E. coli,CHO and other mammalian cells in culture are highly sensitive to the environmentalconditions. The temperature and pH have to be kept within an optimal range(pH: 6.9-7.4 (Burgener and Butler, 2006), T: ⇡37±0.1 degrees Celsius (Zhang,2010)), and a complex medium has to be used to supply carbohydrates, aminoacids, inorganic salts, vitamins, hormones/growth factors, lipids and other nutrientsnecessary for cell growth. Oxygen must be supplied to support cell respiration. Theosmolality of the medium (a measure of the osmoles (Osm) of solute per kilogramof solvent (Osm/kg)), needs to be maintained at approximately 300 mOsm/kg. Thisis to prevent osmotic imbalances between the cell and its surroundings (Burgenerand Butler, 2006). A rough control over pH can be achieved via the addition ofbu↵ers to the cultivation medium; bicarbonate, and potentially HEPES, is used incombination with carbon dioxide supplied to the headspace. Typically, a mixture of95% air and 5% carbon dioxide is used, which is in line with the pCO2 physiologicalrange (Altman and Dittmer, 1971; Dezengotita et al., 1998). The carbon dioxidein the headspace dissolves and is in equilibrium with the bicarbonate bu↵er in themedium.

2.2.7 Cultivation modes

The batch, fed-batch (Xie and Wang, 1994a; Xie and Zhou, 2006) and continuouscultivation modes (e.g. chemostat cultures and perfusion) (Zhang, 2010) are well-established techniques within cultivation technology (Wittmann et al., 2017).

13

Page 32: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

For the large-scale production of biopharmaceuticals using mammalian celllines, the batch or fed-batch modes are the most common (Xie and Wang, 1994a;Xie and Zhou, 2006; Zhang, 2010). In batch mode, cells are seeded in a cultivationmedium after which no additional additions are made. In fed-batch mode, a feedmedium is fed to the cultivation in a strategic manner, primarily to avoid nutrientdepletion.

The timeline of these cultivations can be divided into four phases. The lagphase corresponds to the period of limited growth that begins immediately afterseeding, during which the cells adapt to their new environment. The transitioninto the exponential phase is marked by an acceleration of the growth rate, whicheventually stabilizes at a high and constant rate.

As the number of cells increases rapidly, the nutrient concentrations decrease.In the fed-batch mode, the addition of a concentrated nutrient solution at particularintervals can increase the maximum cell density, and prolong the cultivation (Zhang,2010). Regardless, by-products (primarily lactate and ammonium) begin to accu-mulate, inhibiting growth and promoting death. A decreasing growth rate marksthe transition into the stationary phase, during which growth and death rates are inbalance. Eventually, the cultivation enters the death phase; the death rate exceedsthe growth rate causing a decline in cell number.

In the continuous mode, e.g. chemostat, medium is continuously fed and culturebroth (including cells) removed. The result is a stable environment with a continuoussupply of nutrients and by-product removal. Chemostat culture is seldom used formammalian cells, as slow growth rates can lead to washout e↵ects.

Instead, continuous perfusion (Kompala and Sadettin, 2006) has become in-creasingly popular within the biopharmaceutical industry. This is a variant of thecontinuous mode in which the cells are retained inside or recycled back to thebioreactor, using e.g. filter, sedimentation, centrifugation or acoustic aggregationdevices to separate the cells from the liquid (Kompala and Sadettin, 2006). Eventhough perfusions are associated with high costs (Henry et al., 2008), contaminationrisk, and complex set up and operation, they can last for several months, achievevery high cell densities (Clincke et al., 2013; Zhang et al., 2015; Chotteau et al.,2015) and ultimately generate large amounts of product (Zhang, 2010).

For the present work, a cultivation mode referred to as pseudo-perfusion (orsemi-continuous (Henry et al., 2008)) was chosen. This mode is a simplified versionof the perfusion mode, as the medium renewal is carried out at discrete time-points. This reaps some of the benefits of a perfusion, yet can be carried out usingsimple equipment available in most cell culture laboratories (e.g. small-scale non-instrumented bioreactors, an incubator, a laminar air flow bench, and a centrifuge).

14

Page 33: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.2. Production and process development

Thereby, pseudo-perfusion suits small-scale and parallel screening experiments, inwhich selected process parameters are tested on many di↵erent levels.

2.2.8 Bioreactors

A wide range of bioreactors are available for the cultivation of CHO cells (Zhang,2010). While the original CHO cell lines were adherent, the description here islimited to CHO cells in suspension which dominates in the industry (Zhang, 2010).

Small-scale and non-instrumented bioreactors, e.g. flasks, tubes or multi-wellplates, are used as models of the final large-scale bioreactor. These are kept insideincubators that provide the appropriate temperature and an atmosphere of carbondioxide and oxygen. Loose caps allow for exchange of gas between the culture andthe atmosphere of the incubator. Gentle mixing to ensure a homogenous suspensionand distribution of gases is achieved by placing the flasks on shaking tables. TheTubeSpin bioreactors applied in this thesis belong to this category. A TubeSpinresembles typical centrifuge plastic tube (e.g. 50 mL), but comes with a vented cap.Multiple tubes can be placed in racks, allowing for parallel cultures to be carriedout, e.g. to screen di↵erent media.

The simple bioreactors described in the previous paragraph lack the monitoringand control required in the final production process. A step-wise scale-up towardthe final process is therefore carried out in bioreactors with an increased level ofinstrumentation. pH, pO2 and temperature are monitored using electrodes. pHcontrol is achieved through automated addition of acid (1M HCl) and base (1MNaOH). pO2 control is achieved through controlled inflow of a CO2/O2 gas mixture.Deviations in the temperature is counteracted by automated heating. In addition,shear e↵ects on the cells resulting from primarily agitation and bubble aerationcan be studied (Pörtner et al., 2017); this is to achieve a strategy that minimizesadverse shear and cell damage while still achieving su�cient mixing and gas supply(Aunins and Henzler, 2008).

In the present work, cell cultures were carried out in TubeSpin bioreactors inthe pseudo-perfusion mode. While this set up serves as a model of the continuousperfusions used in industry, an important di↵erence is the lack of control oversome process parameters achieved in larger instrumented systems. Exchange of themedium at discrete time points can lead to significant fluctuations in e.g. pH, dis-solved oxygen, and nutrient and by-product concentrations. From that perspective,an alternative name for pseudo-perfusion is "repeated batch" since the cells experi-ence similar conditions as during the exponential growth phase of batch-cultures(Henry et al., 2008).

15

Page 34: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

2.2.9 Medium development

Changes in the medium composition can have dramatic e↵ects on the growth andproduct yield and/or quality (Zhang, 2010; Xing et al., 2011), but presents a highlychallenging task (Bhambure et al., 2011). Even if medium development can becomplex and time-consuming, it is an opportunity to improve process outcomes.An optimal medium formulation constitutes intellectual property of considerablevalue. Consequently, the composition of most high-performance commercial mediaare proprietary and not publicly available (Zhang, 2010).

Culture media used for mammalian cells contain a high number of components.The nutritional requirements vary between di↵erent cell lines and candidate clones,and may also be influenced by the composition of the biopharmaceutical product(Carrillo-Cocom et al., 2015).

In the past, animal-derived components (e.g. from blood or tissue) were usedin media. The first CHO cell cultures carried out by Puck, Theodore et al. (1958)relied on a defined nutrient solution of amino acids, vitamins, inorganic salts,glucose, and some other components, supplemented with antibiotics and fetal calfserum at 15 %. The potential interference of unknown components and issues withreproducibility made these media inappropriate for some experimental work. Inaddition, the reliance on animal-derived components was a major disadvantage inbiopharmaceutical production due to contamination risks, for which an exampleis Bovine spongiformous encephalopathy (BSE) contamination in sera from cows(Wurm, 2013).

Over the years, considerable e↵orts were made to reduce and finally elim-inate the dependence on chemically undefined and animal-derived components.Nowadays, chemically-defined media free of animal-derived components are com-mercially available. In addition, modern media are frequently protein-free since thisbenefits product purification.

Cell culture media are mixtures of carbohydrates, amino acids, inorganic salts,vitamins, hormones/growth factors, lipids, and bu↵ers. Glucose is the most com-monly used carbohydrate. In addition, the amino acid glutamine serves as a majorenergy source. The availability of vitamins is essential to the cells, and to cellgrowth and productivity. Vitamins function as co-factors for many enzymes. Supp-lementation of lipids can significantly improve the performance of cell cultures, as itreduces the need for lipid biosynthesis, and certain lipids are essential for some celllines. The medium also helps to maintain pH and osmolality within an acceptablerange. The pH is maintained via the use of bu↵ers; i.e. sodium bicarbonate aspreviously mentioned.

16

Page 35: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.2. Production and process development

Medium optimization means solving a multifactorial and combinatorial problem.There is a high number of potential components to choose from, and the combinationof components is important rather than the individual components themselves(Burgener and Butler, 2006). Initially, several di↵erent candidate media, andmixtures of them, can be screened.

The optimal concentration of a specific component can also be determinedby examining the growth response at di↵erent concentrations. However, due tocomponent interaction e↵ects, combinations of multiple components at variedconcentration levels must also be studied. Components with positive e↵ect in othermedia or cultures may in another scenario have little impact, an may become toxicdue to such interactions. The media must also be chemically stable, during storeand in culture.

In depletion analysis, component concentrations in spent medium are analyzedto reveal the nutrient profiles of a specific process. Depleted nutrients shouldbe increased in concentration (Xie and Wang, 1994b; Bibila and Robinson, 1995;Bradley et al., 2010; Sellick et al., 2011), or supplied via feeds, (Bibila and Robinson,1995), and components in excess could potentially be reduced in concentration (Xinget al., 2011).

It is clear that there is a close connection between the development of mediaand the cell metabolism. The medium provides the nutrients processed by themetabolism to support the cell growth and product formation. In that context, anideal medium promotes a metabolism characterized by an e�cient usage of thesubstrates, with limited formation of by-products.

The metabolism of cells in culture can be characterized experimentally bysampling and concentration analysis of important medium components, the cellnumber, the product, and several by-products at consecutive time points. Fromsuch data follow the calculations of the corresponding cell-specific growth andproduction rates (see (2.1) and (2.3), p. 12), and several metabolite uptake/secretionrates. The latter can be calculated as follows for a metabolite i:

qext = µ ·ci � ci,0

Cv �Cv,0, (2.4)

After the rate data have been calculated, mathematical models can be used tointerpret such data, and to guide the design of media and feeds, e.g. using statisticalmethods or flux analysis, which will be described in the next chapter.

17

Page 36: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

2.3 The metabolism of the CHO cell linesRapidly growing CHO cells are characterized by an altered overflow metabolism:high glucose and glutamine uptake rates, and high lactate and ammonium secretionrates (Gódia and Cairó, 2006). In a batch cultivation, glucose and glutamine arerapidly depleted from the medium, while lactate and ammonium accumulate. Thepattern of uptake and secretion of amino acids tend to vary between cell lines, andfor a specific cell line when subjected to di↵erent culture conditions.

In this section metabolic subsystems, frequently referred to in metabolic cellculture modeling, will be described. Figure 2.2 shows an overview of those subsys-tems in the CHO cell metabolism. The reactions of a reaction network applied inPaper III of this thesis will be referred to throughout the section (see Figure 7.1,p. 106).

2.3.1 Overview of cell metabolism

Cell metabolism involves the chemical processes occurring within the cell result-ing in growth, energy production and elimination of waste. Catabolism involvesmetabolic processes in which complex molecules are broken down into simple oneswith the release of energy. Anabolism (also called biosynthesis) involves metabolicprocesses in which complex molecules, e.g. proteins and fats, are synthesized fromsimpler ones.

A metabolic pathway consists of several reactions in series, and most of thosereactions are catalyzed by enzymes. The metabolic flux through a pathway refersto the turnover rate of metabolites through that pathway. This flux is regulated viadi↵erent mechanisms so that current demands are met, e.g. to support biosynthesisand energy generation.

Anabolic pathways lead to the synthesis of complex macromolecules essentialfor life, i.e. nucleic acids (DNA and RNA), proteins, lipids and complex carbo-hydrates. Catabolic pathways lead to the breakdown of complex molecules togenerate energy. In principle, the energy released from catabolic pathways is usedby anabolic pathways. Some metabolic pathways can both produce and utilizeenergy, depending on the current energy need and availability.

The metabolism of CHO cells has been described as flexible (Altamirano et al.,2013); there is an inherent ability to balance the use of di↵erent metabolic subsys-tems to counteract changes in their environment. This metabolism will be coveredin Sections 2.3.2-2.3.11. Here follows an overview of the most important pathwaysand their functions in the cell.

18

Page 37: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.3. The metabolism of the CHO cell lines

• The glycolysis pathway, the TCA-cylce, and the oxidative phosphorylation,break down nutrients and transfer the energy released into energy molecules(e.g. ATP, GTP, NADH, NADPH and FADH2).

– The glycolysis pathway converts glucose, the main source of energy ofliving organisms, into pyruvate. ATP and NADH are generated.

– The pentose-phosphate pathway (PPP) runs parallel to the glycolysis andconverts glucose (via G6P) into pentose sugars and ribose-5-phophate(a precursor for nucleotide synthesis). NADPH is generated.

– The tricarboxylic acid cycle, TCA-cycle, is also known as the Krebscycle or citric acid cycle, and carries out the oxidation of Acetyl-CoAinto carbon dioxide and water. NADH, FADH2 and GTP are generated.

– NADH and FADH2 are used in the oxidative phosphorylation pathway.ATP is generated. Oxygen is the final electron acceptor in aerobicrespiration, and is reduced to water and energy stored as ATP.

• Examples of pathways involved in anabolism/biosynthesis are the synthesesof the cell membrane, DNA and RNA, and proteins.

– The cell membrane consists of lipids: phospholipids, sphingolipids andcholesterol. Phospholipids and sphingolipids are made from fatty acids.

– DNA and RNA are built from purine and pyrimidine nucleotides.– Proteins are built from amino acids.

The biochemical reactions and metabolites of eukaryotes (e.g. protists, fungi,animals and plants) are separated into cellular compartments (Schryer et al., 2009;Wahrheit et al., 2011), making eukaryotic metabolism highly complex. Withinthe cytosol are numerous membrane-bound organelles, i.e. the cell nucleus, themitochondrium, the endoplasmatic reticulum, the Glogi apparatus, the ribosomes,the lysosome, and the peroxisome. Each compartment carries out specializedfunctions, but needs to communicate and co-operate with others via signaling andmetabolite transport (Wahrheit et al., 2011). A metabolic pathway can span severalcompartments, reactions can occur in parallel within di↵erent compartments, andthere can be distinct subpopulations of some organelles (Wahrheit et al., 2011).

2.3.2 Glucose uptake and lactate secretion

A metabolic pathway crucial for understanding mammalian and CHO cell metabolismis the glycolysis, and its relation to lactate metabolism. The high-rate conversion of

19

Page 38: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

Nucleotidesynthesis

PPP

Glycolysis

Lipidsynthesis

NEAAsNEAA

metabolism

Urea cycle

EEAs EAA metabolism

TCA cycle

Biomass synthesis

Urea

NH4

NH4

GLC

GLN

LAC

mABsynthesis

Figure 2.2: An overview of cell metabolism with important metabolic subsystems. Sub-strates (glucose and glutamine) and by-products (lactate and ammonium) important in CHOcells are indicated. The dashed red line indicates the location of the TCA-cycle in themitochondria. GLC: glucose; GLN: glutamine; LAC: lactate; NH4: ammonium, NH4

+.This overview follows the arrangement of the subsystems in Figure 7.1 (p. 106), in whichthe biochemical reactions are visible in the N126 network map from Paper III.

glucose into lactate in fast-growing CHO cells is the result of an ine�cient overflowmetabolism that does provide ATP, but also leads to extensive by-product formation.

Glucose is converted into pyruvate via a number of intermediates (Table 2.1).Two molecules of ATP and two molecules of NADH are obtained. At this point,two main routes for pyruvate are usually considered: either the conversion intolactate, or the entry into the mitochondria for entering the TCA-cycle. Typically, alarge fraction of the pyruvate generated via the glycolysis is converted into lactate.CHO cells operate with aerobic glycolysis (Mulukutla et al., 2010; Altamirano et al.,2013), meaning that this conversion occurs even when oxygen is available. Thislactate production stemming from aerobic glycolysis is a characteristic feature of

20

Page 39: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.3. The metabolism of the CHO cell lines

growing cells in culture (Gódia and Cairó, 2006; Mulukutla et al., 2012), and issimilar to "the Warburg E↵ect" observed in cancer/tumor cells (Warburg, 1956).

Table 2.1: The reactions of the glycolysis in network N126 (v1-v8), and a connection to theTCA-cycle via transport of pyruvate into the mitochondria (v91), a conversion of pyruvateinto AcCoA (v14), which then enters the cycle via v15.

Glycolysisv1 Glcext+ATP! G6P + ADPv2 G6P$ F6Pv3 F6P + ATP! DHAP + GA3P + ADPv4 DHAP$ GA3Pv5 GA3P + ADP + NAD+ $ 3PG + ATP + NADHv6 3PG$ PEPv7 PEP + ADP! Pyr + ATPv8 Pyr + NADH! Lacext + NAD+

A connection to the TCA-cyclev91 Pyr! Pyrmv14 Pyrm + NAD+ ! AcCoAm + CO2,m + NADHv15 AcCoAm + Oxam ! Citm

Di↵erent explanations have been proposed (Gódia and Cairó, 2006). Onesuggests that the conversion of pyruvate into lactate via the lactate dehydrogenaseenzyme is favored to regenerate the co-factor NAD+ (Gódia and Cairó, 2006). Whilethe glycolytic conversion of glucose to pyruvate consumes NAD+, the conversionof pyruvate into lactate catalyzed by the lactate dehydrogenase enzyme regeneratesNAD+, and could thus be necessary to maintain the cytosolic NAD+/NADH bal-ance. Another explanation is a potential limitation in the capacity of the enzymesresponsible for the link between the glycolysis and the TCA-cycle.

The secretion of lactate into the medium can have detrimental e↵ects on the cellculture. High lactate concentrations, ⇡ 35-40 mM, tend to occur in the later stagesof batch and fed-batch cultivations. Lactate accumulation in the medium leads toa decrease in the pH. CO2 production associated with cell growth has a similare↵ect. This can be counteracted by pH control systems as previously described. Theaddition of base (NaOH) to raise the pH to the set point does however increase theosmolality of the medium, which is also problematic (Dezengotita et al., 1998).

Examples of strategies that can reduce lactate accumulation in mammalian cell

21

Page 40: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

cultures are the use of alternative sugars instead of glucose (Altamirano et al., 2006),a controlled feeding of glucose (Gambhir et al., 1999), and various cell engineeringstrategies (Zhou et al., 2011).

2.3.3 Pentose Phosphate Pathway (PPP)

The pentose phosphate pathway (PPP) provides an alternative route for G6P (glucose-6-phosphate), an intermediate generated in the glycolysis (Table 2.2). The role ofthe PPP is mostly anabolic, meaning that it supplies building blocks for biosyntheticprocesses: NADPH, pentoses and ribose-5-phosphate. Ribose-5-phosphate is usedin the nucleotide synthesis (Figures 2.2 and 7.1).

The PPP can be divided into two phases. The first phase is termed oxidativeand generates NADPH for fatty acid synthesis. The second phase is termed non-oxidative and generates six-carbon sugars. The PPP also results in release of carbondioxide.

Table 2.2: The reactions from the PPP in N126 (v9-v13). Metabolites found in the glycolysisare marked in bold. Rl5P, which occurs in v9-v11, is a metabolite that enters the nucleotidesynthesis.

Pentose phosphate pathway (PPP)v9 G6P + 2 NADP+ ! Rl5P + CO2 + 2 NADPH

v10 Rl5P$ R5Pv11 Rl5P$ X5Pv12 R5P + X5P$ F6P + E4Pv13 X5P + E4P$F6P + GA3P

2.3.4 The TCA-cycle

The tricarboxylic acid cycle (TCA-cycle), also known as the Krebs cycle or citricacid cycle, acts as a central hub of the cell metabolism, as it is connected to severalof the other metabolic pathways (Table 2.3). But as the TCA-cycle is primarilylocated in the mitochondria, it is separated from many of the other pathways, andseveral metabolites need to be transported in between.

A key role of the TCA-cycle is in aerobic respiration, to which it supplies co-factors for the energy generation. In terms of the number of energetic co-factors thatcan be produced per carbon, the TCA-cycle is more e�cient than the glycolysis. InCHO cells, despite this di↵erence, only a small fraction of the pyruvate generated in

22

Page 41: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.3. The metabolism of the CHO cell lines

the glycolysis appears to enter the mitochondria for participating in the TCA-cycle(Gódia and Cairó, 2006).

Table 2.3: Reactions from the TCA-cycle in network N126 (v14-v25).

TCA-cyclev14 Pyrm + NAD+ ! AcCoAm + CO2,m + NADHv15 AcCoAm + Oxam ! Citmv16 Citm + NAD(P)+ ! aKGm + CO2,m + NADPHv17 aKGm + NAD+ ! SucCoAm + CO2,m + NADHv18 SucCoAm + GDP$ Sucm + GTPv19 Sucm FAD+$ Fumm + FADH2v20 Fumm $Malmv21 Malm + NAD+ $ Oxam + NADHv22 Cit$ AcCoA + Oxav23 Cit + NAD(P)+ ! aKG + CO2 + NAD(P)Hv24 Fum$Malv25 Mal + NAD+$ Oxa + NADH

The TCA-cycle is closely linked to the metabolism of amino acids, which willbe described in more detail in the next section. As will be seen, the conversionbetween di↵erent amino acids often involve the TCA-cycle intermediates.

2.3.5 Introduction to the amino acid metabolism

Amino acids play a crucial role in the metabolism of mammalian cells. They actas precursors for the proteins and nucleotides important in cell growth, and as themain building blocks of the recombinant protein product. Some amino acids areessential, as they cannot be synthesized by the cells, while other amino acids can besynthesized (Table 2.7). Nevertheless, all the 20 natural amino acids are typicallyprovided in cell culture media to support cell growth and product formation (Xieand Wang, 1996a).

The pattern of uptake/secretion rates for amino acids in cell cultures can beunpredictable (Table 2.9, p. 31). The prevalence of amino acids in the recombinantprotein product can demand for higher consumption and/or synthesis of certainamino acids (Carrillo-Cocom et al., 2015). The pattern also depends on the medium(Wahrheit et al., 2014a; Hagrot et al., 2017) and current process parameters, andcan vary throughout a cultivation as the cell number and extracellular environment

23

Page 42: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

change (Wahrheit et al., 2014a). Increasing the concentration of certain amino acidsappears to protect cells from adverse e↵ects due to e.g. elevated ammonium (Chenand Harcum, 2005), carbon dioxide or osmolality (Vivian et al., 2002).

2.3.6 Essential and non-essential amino acids

The amino acids are usually divided into: essential; non-essential; and conditionallyessential. The essential amino acids (EAAs) must be supplied in the culture medium,as they cannot be synthesized by the cells. Essential amino acids include histidine,isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan andvaline. It is expected that growing CHO cells would consume EAAs, as theseare building blocks of proteins and product. EAAs can also be degraded viaEAA catabolism (Table 2.4). Their degradation routes replenish the TCA-cyclevia AcCoA and SucCoA. Ammonium, carbon dioxide, and glutamate are majorproducts of this metabolism, and aKG frequently occurs as a substrate.

The NEAAs of the mammalian cell metabolism include alanine, asparagine,aspartate, glutamate, glutamine, glycine, proline, and serine. These can be synthe-sized by the cells, and can thereby be both taken up and secreted. Isotopic tracerstudies have revealed that in CHO cells, the degradation and synthesis of someof the non-essential amino acids appear to occur simultaneously (Wahrheit et al.,2014b; Ahn and Antoniewicz, 2012; Deshpande et al., 2009), and that some of thoseamino acids are reversibly exchanged with the surrounding medium (Nicolae et al.,2014).

The conditionally essential amino acids, CEAAs, can be synthesized by thecells; however, their syntheses do depend on the availability of certain other aminoacids. The following three examples are given by Gódia and Cairó (2006) forthe general mammalian cell line metabolism: tyrosine can be synthesized fromphenylalanine; cysteine can be synthesized from methionine; and arginine can besynthesized by the arginase enzyme. Thereby, the essentiality of these amino acidsis situation-dependent.

Finally, certain amino acids are essential for specific cell lines (auxotrophy).The CHO-K1 cell lines are proline-auxotrophic (Kao and Puck, 1967; Wurm, 2013)and thereby requires proline in the culture medium. Unless transfected with vectors,the GS-CHO cell lines are glutamine-auxotrophic, and DHFR-deficient cell linesCHO-DG44 and CHO-DXB11 also requires glycine for growth. A more recentcomparison of CHO cell lines, carried out during the development of large-scalemetabolic models, indicated that the CHO-K1 cell line are actually auxotroph alsofor the amino acids arginine and cysteine (Hefzi et al., 2016).

24

Page 43: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.3. The metabolism of the CHO cell lines

Table 2.4: The reactions of the EAA metabolism in network N126 (v46-v63).

Essential amino acid metabolismv46 Thr + NAD+ ! Gly + AcCoAm + NADHv47 Thr! aKbut + NH4

+

v48 aKbut + NAD+ ! PropCoA + CO2 + NADHv49 PropCoA + CO2 + ATP! SucCoAm + ADPv50 Trp! Ala + aKad + 2 CO2v51 Lys + 2 aKG + 3 NAD(P)+ + FAD! 2 Glu + aKad + 3 NAD(P)H + FADH2v52 aKad + 2 NAD+ ! AcetoacCoA + 2 CO2 + 2 NADHv53 AcetoacCoA! 2 AcCoAmv54 Val + aKG + 3 NAD+ + FAD

! Glu + PropCoA + 2 CO2 + 3 NADH + FADH2v55 Ile + aKG + 2 NAD+ + FAD

! Glu + PropCoA + CO2 + AcCoAm + 3 NADH + FADH2v56 Leu + aKG + ATP + NAD+ + FAD

! Glu + Acetoac + AcCoAm + ADP + NADH + FADH2v57 Acetoac + SucCoAm ! AcetoacCoA+ Sucmv58 Phe + NADH! Tyr + NAD+v59 Tyr + aKG! Glu + Fum + Acetoac + CO2v60 Met + Ser + ATP! Cys + aKbut + NH4

+ + AMPv61 Cys! Pyr + NH4

+

v62 Arg! Orn + Ureav63 His! Glu + NH4

+

2.3.7 Glutamine uptake and ammonium secretion

The amino acid glutamine is of particular interest in mammalian cell culture, sinceit is usually consumed at high rates which leads to overflow metabolism; glu-tamine uptake and subsequent catabolism exceed what would be needed for purelybiosynthetic requirements (Altamirano et al., 2013). The result is an extensiveammonium formation and secretion into the medium.

Ammonium is a potentially toxic and inhibitory by-product in CHO cell cultures(Chen and Harcum, 2005). It can inhibit cell growth and negatively a↵ect theproductivity and quality of the recombinant product (e.g. on the glycosylationprofile). Ammonium generation is closely linked to the metabolism of amino acids,and in particular to the glutamine uptake and degradation routes referred to as theglutaminolysis.

In one route of the glutaminolysis, glutamine is first converted into glutamate,

25

Page 44: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

Table 2.5: The reactions of the NEAA metabolism in network N126 (v30-v45).

Non-essential amino acid metabolismv30 Pyr + Glu$ aKG + Alav31 Pyrm + Glum $ aKGm + Alamv32 Oxa + Glu$ aKG + Aspv33 Oxam + Glum $ aKGm + Aspmv34 Asn! Asp + NH4

+

v35 Asn + Glu$ Gln + Aspv36 Gln$ Glu + NH4

+

v37 Glu + NAD(P)+ $ aKG + NH4+ + NAD(P)H

v38 Glum NAD(P)+ $ aKGm + NH4m + NAD(P)Hv39 3PG + Glu + NAD+! aKG + Ser + NADHv40 Ser! Pyr + NH4

+

v41 Ser$ Glyv42 Gly$ CO2,m + NH4

+

v43 aKG + Orn$ Glu + Glu�SAv44 Pro! Glu�SAv45 Glu�SA$ Glu

and then into aKG to replenish the TCA-cycle. Each step of this route generatesammonium, leading to formation of two moles of ammonium ions per mole ofglutamine.

A second route converts the glutamate and pyruvate into aKG and alanine viaan alanine transaminase enzyme. This conversion does not generate ammonium.In this way, alanine serves as a nitrogen sink, which provides an alternative routewith reduced ammonium formation. This can explain why alanine is often secretedas a by-product when CHO cells are cultured in glutamine-containing media (seeTable 2.9).

In a third route, aspartate transaminase converts oxaloacetate and glutamateinto aspartate and aKG. Again, this conversion does not generate ammonium, andaspartate can thereby serve as a nitrogen sink similar to alanine.

26

Page 45: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.3. The metabolism of the CHO cell lines

Pyruvate

aKG

Succinate

Isocitrate

MalateMalate

Pyruvate

Oxaloacetate

PEP

Glu GLN

Ala Asp

asp TA

ala TA

GDH

MEME

PEPck

MDH MDH

PC

LAC

GLC

TCA

Glycolysis

Glutaminolysis

Mitochondrion

GS

Acetyl-CoA

A

B

C

Lipid synthesis

Succinyl-CoAFumarate

CitrateOxaloacetate

Cytosol

NH4+ NH4+

Figure 2.3: Routes of the glutaminolysis and the connection between the glutaminolysisand the glycolysis. A: Degradation of glutamine into aKG via glutamine synthetase (GS)and glutamate dehydrogenase (GDH). B: Alternative routes via transaminase enzymes,alanine transaminase (ala TA) and aspartate transaminase (asp TA). C: Conversion ofmalate/oxaloacetate into pyruvate; for enzymes see the text and Table 2.6. Note that citrateis extracted from the cycle to supply lipid synthesis. This figure is based on illustrations byGódia and Cairó (2006), specifically Figure 3 and 4.

There is a connection between the glycolysis and the glutaminolysis (Figure 2.3).aKG derived from glutamine enters the TCA-cycle, and is converted into malate viathe lower half of the cycle. The frequent route to account for this overflow in theTCA-cycle is via the malate shunt (Gódia and Cairó, 2006); malate is exported outof the mitochondrion, and then converted into pyruvate via a cytosolic malic enzyme(ME) (Niklas et al., 2011). This connects the glutaminolysis to the glycolysis, andpyruvate derived from glutamine can be converted into lactate. The degradation ofglutamine is thereby linked to the lactate secretion.

Other routes connecting malate to pyruvate are possible, and it can be di�cult toseparate them experimentally (Niklas and Heinzle, 2011), see Table 2.6. In network

27

Page 46: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

Table 2.6: The anaplerotic reactions in network N126 (v26-v29). ME: malic enzyme. PC:pyruvate carboxylase. PEPck: phosphoenolpyruvate carboxykinase.

Anaplerotic reactionsME v26 Malm + NAD(P)+ $ Pyrm + CO2 + NAD(P)HPC v27 Oxam + ADP$ Pyrm + CO2 + ATPPEPck v28 PEP + CO2 + GDP$ Oxa + GTPME v29 Mal + NAD(P)+ $ Pyr + CO2 + NAD(P)H

N126, there is a mitochondrial version of ME that can convert malate into pyruvate,within the mitochondria. Another route involves oxaloacetate. Oxaloacetate isobtained from malate in the TCA-cycle, but this reaction can also take place in thecytosol via malate dehydrogenase (MDH). Oxaloacetate can then be converted intoPEP, an intermediate of the glycolysis, by the phosphoenolpyruvate carboxykinase(PEPck) enzyme (Niklas et al., 2011). Finally, the enzyme pyruvate carboxylase(PC) can perform the opposite conversion of pyruvate into oxaloacetate within themitochondria.

2.3.8 General trends of amino acid uptake/secretion patterns

Gódia and Cairó (2006) divides amino acids into three groups based on the generaltrend of consumption/production in mammalian cell cultures. Valine, isoleucine,leucine, lysine and cysteine are usually consumed rapidly. Threonine, arginine,phenylalanine, serine, histidine, methionine and glycine usually remain constant orare consumed in fair amounts. Alanine, proline, asparagine and glutamate are typi-cally produced. Table 2.9 lists the amino acids and the pattern of uptake/secretiondescribed by (Gódia and Cairó, 2006), and as reported in some literature for CHOcells (Gódia and Cairó, 2006; Zamorano et al., 2010; Wahrheit et al., 2014a). Thepatterns observed in the present work are also indicated (the generation of thosedata are presented in Chapter 4).

2.3.9 Amino acid synthesis

NEAAs can be synthesized within the cell. In Table 2.5, ten of the sixteen reactionsin the NEAA metabolism are reversible, and some of the NEAAs can be synthesizedvia the degradation of EAAs in the EAA metabolism.

28

Page 47: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.3. The metabolism of the CHO cell lines

As shown in Table 2.4, glutamate is produced in six reactions of the EAAmetabolism; i.e. in the degradations of lysine (v51), valine (v54), isoleucine (v55),leucine (v56), tyrosine (v59), and histidine (v63). Glycine and alanine are produced indegradation of threonine (v46) and tryptophan (v50), respectively. Serine is consumedin the degradation of methionine (v60).

As can be seen in Table 2.7, glutamate is a highly connected metabolite withinthe amino acid synthesis and metabolism. Glutamate can be obtained from ten otheramino acids. Noteworthy is that aspartate can be obtained from asparagine, andserine and glycine are interconvertible, as are glutamine and asparagine.

Table 2.7: Synthesis of amino acids. Proline synthesis is not represented as CHO-K1 cellsare definitely auxotrophs for this amino acid.

Can be obtained fromAmino acid NEAAs (C)EAAs

NEAAsAla Glu TrpAsn GlnAsp Glu, AsnGlu Ala, Asp, Gln, Pro Lys, Val, Ile, Leu, Tyr, HisGln Glu, AsnGly Ser ThrSer Gly

CEAAsCys MetTyr PheArg Asp, Glu

2.3.10 Balancing ammonium

The metabolism of amino acids contributes to ammonium formation, and to am-monium consumption. At high ammonium, the metabolism can adapt; ammoniumrelease is reduced and ammonium fixation supported via increased production andsecretion, or decreased uptake and degradation, of specific amino acids (Wahrheitet al., 2014a). As seen in Table 2.4, four reactions in the EAA metabolism generateammonium, i.e. degradations of threonine (v47), methionine (v60), cysteine (v61) and

29

Page 48: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

histidine (v63). And as seen in Table 2.5, four out of the five ammonium–generatingreactions in the NEAA metabolism are reversible in network N126.

2.3.11 Urea cycle

The metabolism of urea in the urea cycle is connected to the detoxification ofammonium, and to amino acids glutamate and arginine (Table 2.8). In animals,the urea cycle contributes to ammonium detoxification, primarily in the liver, byconverting ammonium into urea that is secreted. The activity of the cycle in CHOcell lines has been uncertain. While mammalian tissue is ureotelic (Bonarius et al.,1996), it is usually assumed that the urea cycle is not active in tumor cells (Colemanand Lavietes, 1981; Bonarius et al., 1996; Martens, 2007), industrial mammaliancell lines (Martens, 2007) or CHO cells (Altamirano et al., 2013). Zamorano et al.(2010) reported that small amounts of urea could be detected during cultures ofa CHO cell line. In a more recent study, the concentration of urea was analyzedduring cultures of a CHO-K1 cell line under varied availability of glutamine; in thatcase, no urea was detected (Wahrheit et al., 2014a).

Table 2.8: The reactions of the urea cycle in network N126 (v64-v66).

Urea cyclev64 Orn + NH4

+m + 1 CO2,m + 2 ATP! 1 Cln + 2 ADP

v65 Asp + Cln + ATP! Argsucc + AMPv66 Argsucc! Fum + Arg

As pointed out by Martens (2007), it is possible that products of the urea cycleother than urea could be secreted during a culture (e.g. citrulline and/or ornithine)(Martens, 2007). Furthermore, Park et al. (2000) achieved a reduced ammoniumaccumulation by forcing permanent expression of selected urea cycle enzymes (Parket al., 2000), and pointed out that certain co-factors were needed for the activationof some of those urea cycle enzymes (e.g. ATP, acetylglutamate and ornithine).

30

Page 49: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.3. The metabolism of the CHO cell lines

Table 2.9: Patterns of amino acid uptake/secretion in mammalian cells and CHO cells.E: essential, NE: non-essential, CE: conditionally essential. +: secretion, �: uptake, c:constant, +/�: varying between uptake and secretion, d: depleted. NA: measurement notavailable. General trend refers to the one described by Gódia and Cairó (2006).

Amino acid Req

uire

men

t

Gen

eral

trend

CH

O-3

20ba

tcha

CH

O-K

1ba

tch/

fed-

batc

hb

CH

O-K

1ba

tch/

fed-

batc

hc

CH

OD

UK

Xre

peat

edba

tchd

CH

OD

UK

Xre

peat

edba

tche

CH

O-K

1ps

eudo

-per

fusi

onf

Alanine Ala A NE + + + + + + +

Aspartate Asp D NE + � +/� +/� + + +/�Asparagine Asn N NE + � � � � + �Cysteine Cys C CE + NA � � NA NA �Glutamate Glu E NE + + +/� +/� + + +

Glutamine Gln Q NE � � �/d +/� � � �Glycine Gly G NE �/c + +/�/c +/� + + +/�Proline Pro P NE + � � � NA NA �Serine Ser S NE �/c � � � � � +/�/cTyrosine Tyr Y CE � � � � � � �Arginine Arg R E/CE �/c � � � � � NAHistidine His H E �/c NA/- � � � � NA/�Isoleucine Ile I E �� � � � � � �Leucine Leu L E �� � � � � � �Lysine Lys K E �� � � � � � �Methionine Met M E �/c � � � � � �Phenylalanine Phe F E �/c � � � �Threonine Thr T E �/c � � � � � �Tryptophan Trp W E � NA � � � � �Valine Val V E � � � � � � �a CHO-320 cells in batch (Zamorano et al., 2010).b CHO-K1 cells in glutamine-supplemented medium at di↵erent glutamineconcentrations in either batch or fed-batch mode cultures (Wahrheit et al., 2014a).c CHO-K1 cells in glutamine-free medium in either batch or fed-batch cultures(Wahrheit et al., 2014a).d CHODUKX with asparagine in repeated three-day batch cultures (refers to the twofirst repetitions) (Seewöster and Lehmann, 1995).e CHODUKX without asparagine in repeated three-day batch cultures (refers to the thirdrepetition) (Seewöster and Lehmann, 1995) .f CHO-K1 cells in media with varied amino acid availability in parallellpseudo-perfusion mode cultures (this thesis).

31

Page 50: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2. Biopharmaceutical production

2.4 Future strategies for process optimization withmetabolic models

Process optimization, here relating to the upstream process and in particular to thecultivation, is the action of adjusting the conditions and the process parameters insuch a way that the best possible process outcome is obtained. The best outcomecould depend on multiple objectives, e.g., a high amount of final product, a highproduct quality, and an overall process cost-e↵ectiveness.

2.4.1 A framework for optimization strategies

In their review, Bhambure et al. (2011) divides process optimization strategies intofour types: A, B, C, and D. Strategy A is one used historically for the productionof biopharmaceuticals and entails experience, empirical knowledge, and heuristicapproaches such as rule-of-thumb and intuitive reasoning. Strategy B entails high-throughput experiments based on e.g. robotic equipment, liquid handling devices,sensitive detectors, and data processing and control softwares. Strategy C involvesalgorithms and optimization tools. Finally, Strategy D encompasses hybrid strategiesobtained by combining two or three of A–C. In this thesis, adopting a hybrid strategyappears most interesting, as there are clearly synergistic e↵ects to be gained.

In a hybrid strategy, simply put, A could be said to be of human nature, B is arobotic system, and C a computational program. A includes our acquired knowledgeand experience, books, databases etc., our ability to plan and carry out experiments,interpret results, plan new experiments and generate new knowledge. But there areclearly cognitive limitations of the human mind, as well as limitations in our abilityto e�ciently and accurately perform repetitive and parallel manual tasks. Here,advanced computational programs (C) can compensate for our cognitive limitations,and robotic systems (B) can reduce our dependence on manual labor, and the risk ofhuman error.

To provide an example from medium optimization, a typical experiment to selectpromising candidates is to culture the cell line in di↵erent medium compositionsand register the growth rate, productivity, and product quality. High-throughput(B) increases the number of medium compositions that can be tested for a specificcell line within a reasonable time frame. Algorithms and optimization tools (C)can support the experimental design, the analysis and interpretation of the data, theextrapolation within and beyond the experimental range, and the optimization ofmedia compositions and feeding strategies.

32

Page 51: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

2.4. Future strategies for process optimization with metabolic models

2.4.2 A vision of the future

In the future, robots and computers within biopharmaceutical production mightevolve toward artificially intelligent systems (AI), with abilities such as learning,planning, decision-making, and self-programming. Progress within this area largelydepends on our ability to translate our knowledge, experience and objectives intoinstructions that can be interpreted by our computers and robotic systems, andenable them to provide us with relevant outputs.

For medium development applications, the standpoint taken in this thesis is thatthe synergy between human, robot, and computer will benefit from metabolic mod-els, representing the connection between the medium composition, the underlyingmetabolism, and the resulting process outcome. Below are tasks that the type ofmodels considered in this thesis could hopefully perform, in the future:

1. Interpret culture data obtained in medium screening experiments throughadvanced modeling of the underlying metabolism.

2. Simulate growth rates, productivity and metabolism as functions of di↵erentmedium compositions in order to identify candidates for experimental testing.

3. Identify an optimal medium composition for a particular cell line, product,and production process.

The focus in the next chapter is on useful theory and methods for the generationof such models.

33

Page 52: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work
Page 53: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Chapter 3

Mathematical modeling of cells

"...if validly constructed, a metabolic model is the key to a vast variety ofproperties that cannot be accessed by intuitive reasoning alone"

(Steuer and Junker, 2009)

Figure 3.1 shows a drawing of a general model M, in this case representing acell population producing a biopharmaceutical. The complex cell described in thepervious chapter has been simplified to a box, with inputs and outputs representedby arrows.

McAcBcCpHTemp.

Cell density and growth (Cv, µ)Product secretion (cmAb, qmAb)Substrate uptake (e.g. cGlc, cGln, qGlc, qGln)By-product secretion (e.g. cLac, cNH+4 , qLac, qNH+4 )Glycosylation profile

Figure 3.1: A general macroscopic/black-box model M of a cell population.

Inputs could be the concentrations of extracellular components present in themedium (here cA, cB, and cC for components A, B, and C), potentially complementedby physical parameters such as the pH and/or temperature. The given outputs are: theresulting cell density (Cv) and growth rate (µ); the product secretion (cmAb and qmAb)and quality (here the glycosylation profile); and the metabolic behavior, meaningthe uptake/secretion of selected extracellular metabolites from/to the medium (e.g.cGlc, qGlc, cLac, and qLac). The type of model depicted in Figure 3.1 could be

35

Page 54: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

referred to as a macroscopic model (Ben Yahia et al., 2015), or a black-box model(Bunge, 1963), in which variations in the observable inputs produce a response inthe observable outputs, or in the macroscopic behavior of the cells.

Any model M is a simplified representation of a real-world target T . When itcomes to cell-based biological systems, T is inherently complex. As a consequence,models of cells are always extensively simplified in relation to their real-worldtarget (Bailey, 1998). Despite advancements in the characterization of biologicalsystems, this situation will likely persist, at least within a foreseeable future. Howto generate a model M and what simplifications that can be justified will dependon what the model is to be used for. To generate M for mammalian cell cultures,kinetic macroscopic modeling has been proposed in several papers (Table 3.6), andthe same approach is considered in the present thesis.

In the next section, this chapter begins with a discussion about simplifications,and the ideals that guide the development of modeling methodology and generationof biological models (Section 3.1). This is followed by a description of modelclassifications relevant to the modeling of cell populations (Section 3.2). After this,focus is given to the concepts and methods relevant to arrive at and further developmacroscopic kinetic models (Sections 3.3-3.5). Finally, the chapter ends with thederivation of a macroscopic kinetic model (Section 3.7), which sets the stage for thepresent investigation of this thesis.

3.1 Idealization and guiding ideals in the modelconstruction process

A model is a distorted version of its target, and the process of distortion is referredto as idealization. A guiding ideal is a goal that governs the model constructionprocess. An awareness of idealization–types and guiding ideals can be helpfulin model construction, as it provides a systematic framework against which toevaluate and compare various models and proposed methodologies. In a paperpublished in the Journal of Philosophy, Weisberg (2007) outlines three types ofidealization: Galilean, minimalist, and multiple models idealization (MMI); theseare summarized in Table 3.1. Weisberg (2007) also introduces the five guiding idealslisted in Table 3.2: completeness, simplicity, 1-causal, Maxout, and generality.

The completeness ideal aims toward a true and highly accurate representationof the complex model target, incorporating all the information currently available.This ideal can never be fully realized, but can still be a primary goal. Galileanidealization is driven by this ideal, but is pragmatic: simplifications are accepted mo-

36

Page 55: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.1. Idealization and guiding ideals in the model construction process

Table 3.1: The three types of idealization suggested by Weisberg (2007) that enable themodeling of complex model targets.

Galilean Strives for a complete, accurate and precise model. Sim-plifies to achieve a tractable problem, based on pragmaticjustification. Assumes future de-idealization, and is sensi-tive to the current state of science and technology.

Minimalist Includes those factors that make a di↵erence to the occur-rence and character of the phenomenon under study.

Multiplemodelsidealization(MMI)

Builds multiple related but incompatible models, justifiedby the idea of tradeo↵s. Does not expect a single bestmodel, but recognizes that multiple models are requiredto satisfy our scientific needs.

Table 3.2: Five of the guiding ideals introduced by Weisberg (2007).

Completeness Include every property of the target and the external fac-tors.

Simplicity Include as little as possible to achieve a qualitative predic-tion of the phenomenon.

1-Causal Include only the core or primary causal factors that ac-count for the phenomenon of interest, that is, the "factorsthat make a di↵erence".

MaxOut Values predictive power. Maximize the precision andaccuracy of the model’s output

Generality Increase the number of actual targets (a-general) and pos-sible targets (p-general).

mentarily, but will be removed once more information, theories, and computationalpower become available (Weisberg, 2007). In biological modeling, striving forcompleteness tends to be time-consuming and requires extensive resources. As willbe seen in Section 3.6 on kinetic modeling, the result is often a complex model, forwhich calculations are di�cult, and for which crucial information is still missing.

As a contrast, the simplicity ideal embraces a simplified representation of themodel target. Simple models are generally easy to set up and calculate, and are goodstarting points for testing new ideas, for the development of new methodology, andeventually more complex models. Simple models are also useful in a pedagogical

37

Page 56: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

context, and can help build intuition about the behavior of the system (Weisberg,2007). From the perspective of biopharmaceutical industry, modeling applicationsthat are straightforward and user-friendly are attractive, as they can be adopted byemployees with limited expertise. Simplicity could mean that constraints in timeand costs can be met1.

The 1-causal ideal prioritizes the inclusion of the causal factors that account forthe phenomena under study. This is in line with practice of minimalist idealization.A minimal model could be simple, but is not necessarily so (Weisberg, 2007); thereis no restriction in terms of complexity per se. The 1-causal ideal can be usefulwhen the purpose of the model is to explain and to improve our understanding abouta system’s behavior; it highlights the essential properties, leaving out extraneousdetails.

The Maxout ideal values predictive power, i.e. to maximize the precision andaccuracy of the model’s output. It does not say how to achieve this (Weisberg,2007). Black-box models, which will be introduced in Section 3.2.3, can have greatpredictive power but they do not necessarily explain the system behavior (Bunge,1963). Model selection can be carried out guided by this ideal; a best model, theone with the best fit to data, is chosen from a set of alternative models, and a newbest model is identified as more data come in (Cawley and Talbot, 2010).

The generality ideal strives to increase the number of targets that a particularmodel captures. Certain models can accurately simulate the behavior of cells underspecific conditions, e.g., models developed for the monitoring and control of adefined manufacturing bioprocess need to be valid with high precision within anarrow operating range (Hodgson et al., 2004). The same model may however failto simulate or predict the behavior of the system when culture conditions change,and would thus possess a low degree of generality from that perspective. Whenthe purpose is prediction and optimization, the model must encompass a range ofdi↵erent input parameter values and have an ability to simulate accurate outputs,even outside an experimental range. If this is the purpose of the model, generalitycan become an important guiding ideal.

Clearly, the guiding ideals are not always compatible. There must thereforebe tradeo↵, balancing the ideals against each other. MMI emphasizes that for themodeling of complex phenomena, such as those observed in biological systems,multiple models are required to "satisfy our scientific needs". No single model canhave all desired properties to the highest magnitude (Levins, 1966; Weisberg, 2007).

1 These aspects were stressed by an industry representative at one of my conferenceposter presentations.

38

Page 57: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.2. Model classifications

Naturally, models that prioritize ideals di↵erently will be found in the researchliterature, and the tradeo↵ might change throughout phases of model development.

3.2 Model classificationsMathematical models of cells can be classified into di↵erent categories. Some ofthem, which are frequently referred to will be described in this section to provide awider context for the type of models applied in the present work.

3.2.1 Probabilistic/stochastic versus deterministic models

Probabilistic/stochastic models describe a small number of individual cells ormolecules (Székely and Burrage, 2014), as would be relevant at microscopic scales(Kirkilionis, 2010). In deterministic models, average behavior of the cell pop-ulation is described rather than that of individual cells; growth, production andconsumption are modeled as continuous processes (Tsuchiya et al., 1966; Székelyand Burrage, 2014). Deterministic models tend to be more intuitive (Kirkilionis,2010), and suit the modeling of large cell populations best (Ben Yahia et al., 2015);as in the present work, measurements referring to the properties of individual cellsor molecules are typically not available. All models discussed in this thesis aretherefore deterministic.

3.2.2 A systematic framework for cell population modeling

Table 3.3 illustrates the systematic framework for the classification of mathematicalmodels of fermentation processes, established by Fredrickson et al. (1970). Thisframework has had a large influence on the way biochemical engineers think aboutcells (Fredrickson et al., 1970; Bailey, 1998). It defines four model classes based onthe combination of an either unstructured or structured, with an either unsegregatedor segregated point of view. Within the framework, the unstructured unsegregatedclass is the most idealized, and the structured segregated the most realistic (Bailey,1998).

Unstructured models idealize the cell population as one single component.Structured models describes the cell in more detail, e.g. by dividing the model intoan extracellular and intracellular part; an internal structure of multiple componentsand reactions is accounted for, potentially in combination with relevant transportand enzyme kinetics (Sanderson et al., 1996).

39

Page 58: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

Table 3.3: A systematic framework for the classification of cell population models, basedon Figure 1 in the paper by Bailey (1998).

Unstructured StructuredUnsegregated An average one-component cell. An average multi-component cell.

Segregated A heterogenous population ofone-component cells.

A heterogenous population ofmulti-component cells.

Unsegregated models idealize the cells as a homogeneous population. In con-trast, segregated models account for distinct subpopulations within the culture(Fredrickson et al., 1970; Bailey, 1998). Cell population heterogeneity can berelevant, e.g. to model the proportion of cells in di↵erent phases of the cell cycle(i.e. G1, S and G2/M), or the increasing proportion of apoptotic cells and dead cellsin some cultures (Zeng and Bi, 2006; Naderi et al., 2011). In the present work, thecell populations in the pseudo-perfusion cultures will be idealized as homogenous.

3.2.3 Black-box models"A black box is a fiction representing a set of concrete systems into whichstimuli S impinge and out of which reactions R emerge. The constitution andstructure of the box are altogether irrelevant to the approach under considera-tion, which is purely external or phenomenological. In other words, only thebehavior of the system will be accounted for."

Bunge (1963), p. 346.

Black-box theory idealizes a real world system, i.e. an open system in which thereis a boundary between the system and its surroundings, to a black box. Input andoutput flows represent exchanges between the system and its surroundings. The

40

Page 59: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.2. Model classifications

system is hidden to the observer; only stimuli (inputs) and responses to those stimuli(outputs) can be observed. In the case of an experiment, the observer is in control ofand can vary the inputs, and records the output response. Afterwards, a model isderived from those inputs/outputs.

A pure black-box model of a cell population would belong to the unstructuredunsegregated model class; this is because the internal structure of the cells, orthe existence of subpopulations, are both based on knowledge about the system’sinternal organization. In a pure black-box model, the relation between the inputs andthe outputs is inferred only from the data, and modeled via a suitable mathematicalrelationship, e.g. linear or quadratic polynomial functions. This can also be referredto as data-driven, or empirical/descriptive modeling.

A good example of empirical modeling often applied in engineering is designof experiments (DoE); DoE is a systematic framework for experimental designand predictive modeling of multiple factors and interaction e↵ects (Ryan, 2011).The method highlights correlations between inputs and outputs, and can guide auser toward optimal conditions. However, the fact that the biological properties areignored may impose a limitation on the model scope, and on the model’s explanatorypower.

It could be argued that macroscopic cell culture models are of black-box–type,since macroscopic refers to what can be observed. In practice however, knowledgeabout the organization of the system is often available to the observer, and willinfluence the model construction to some degree. In that case, the model can bereferred to as of grey-box–type. The situation which will be relevant in the presentwork is one in which measurements are limited to the extracellular components, butsome knowledge about the internal structure of the cells is available, and largelyinfluences the formulation of the model equations.

3.2.4 Mechanistic and structured models

In a mechanistic model, the underlying biological mechanisms that give rise tothe observed phenomena are emphasized to provide a more complete and accuratedescription. Biological properties of the target system are introduced to generate astructured model.

The information that characterizes relevant biological systems has increasedsignificantly over the years, and so has the availability of powerful computationaltools (Sanderson et al., 1996). A common standpoint is that mechanistic modelsbased on a priori biochemical information (Kirkilionis, 2010) have the best po-tential for improving our understanding of cellular behavior, and for prediction

41

Page 60: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

and optimization tasks (Sanderson et al., 1996; Hodgson et al., 2004; Zeng and Bi,2006). For these reasons, unstructured models now appear to be something of thepast (Sanderson et al., 1996). A metabolic reaction network is a central componentof a structured metabolic model, and of the reaction network-based macroscopickinetic models developed in this thesis; this is therefore the topic of the next section.

3.3 Introduction to metabolic reaction networks

3.3.1 Representation of metabolism

A metabolic reaction network consists of two main parts: (i) a set of metabolites;and (ii) a set of reactions interconnecting the metabolites to each other, including thestoichiometric ratios associated with these interconversions. The metabolic networkcan be visualized as a two-dimensional node graph in which the metabolites arenodes interconnected by the reactions in the form of arrows. One-headed arrowsrepresent irreversible reactions, while double-headed arrows represent reversiblereactions.

The metabolic network is represented by a stoichiometric matrix A:

A =

0BBBBBBBBBBBBBBB@

a1,1 a1,2 · · · a1,na2,1 a2,2 · · · a2,n...

.... . .

...am,1 am,2 · · · am,n

1CCCCCCCCCCCCCCCA. (3.1)

Each row in A corresponds to a metabolite, and each column to a reaction. Thenon-zero entries in the matrix are the stoichiometric coe�cients, indicating theinvolvement and stoichiometric contribution of the i:th metabolite in the j:th reaction.Substrates are indicated by negative coe�cients (ai, j < 0), and products by positivecoe�cients (ai, j > 0). Zero (ai, j = 0) means that the metabolite is not involved inthat particular reaction (neither as a substrate nor product). Ideally, the network isstoichiometrically balanced; in each reaction the number of atoms of each elementconsumed equals the number produced.

The stoichiometry represented in A can be seen as an invariant property of thebiological system, independent of physiological environmental conditions (Steuerand Junker, 2009) and of time (Llaneras and Picó, 2008). Llaneras and Picó(2008) refers to this stoichiometry as stoichiometric non-adjustable constraints.This information about the modeled system can be considered relatively well-established (Sanderson et al., 1996); it can be obtained from e.g. databases such as

42

Page 61: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.3. Introduction to metabolic reaction networks

KEGG pathways (Kanehisa and Goto, 2000; Kanehisa et al., 2014), or biochemistrytextbooks (Nelson and Cox, 2005).

The reversibility can be indicated in a vector; this vector is of same length as thenumber of reactions, and contains zeros and ones. In the present work, zeros indicatereversibility, and ones indicate irreversibility. An example of an irreversibility vectorof a network with seven reactions could be:

virrev =⇣

1 1 0 1 1 1 1⌘, (3.2)

In this example, the third reaction is reversible and the others are irreversible. Thesame information can be indicated via bounds on each reaction flux v j. The flux isconstrained to a minimum and a maximum value, [vmin, j vmax, j]. Setting vmin, j = 0and vmax, j � 0 means that the reaction is restricted to net fluxes in the forwarddirection.

Metabolites in a simple metabolic network are divided between an extracellularand an intracellular space. For suspension cells, as the ones studied in this thesis,the extracellular space is their outside environment, the cultivation medium, fromwhich the cells take up substrates and into which they secrete products. Theintracellular space is the interior of the cells, and the barrier separating the twospaces is the cell membrane. Metabolic networks can also represent one or several ofthe compartments located within the intracellular space, typically the mitochondria.

Each metabolite of a network is either extracellular, belonging to the set Mext,or intracellular, belonging to the set Mint. Based on this division, the stoichiometricmatrix is split into an extracellular stoichiometric matrix Aext and an intracellularstoichiometric matrix Aint by separation of the rows in A. To define the transportof metabolites between the two spaces, transport reactions (also called exchangereactions) are added to the network by the addition of new columns to A. Additionalcompartments within the intracellular space can be accounted for using the sameprinciple: a separation of metabolite pools and the addition of adequate transportreactions (Sanderson et al., 1996). A common situation, as in the present work, isthat the metabolites in Mext are accessible for concentration analysis. The analysisof the intracellular metabolite concentrations is less straightforward and generallyrequires cell disruption (Seewöster and Lehmann, 1995).

3.3.2 Design of metabolic networks

The metabolism of living cells, including the metabolism of CHO cells, is highlycomplex and consists of thousands of reactions. Many of those reactions are knownand have been organized into metabolic subsystems and metabolic pathways, for

43

Page 62: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

which detailed information is available in biochemistry textbooks (Nelson and Cox,2005) and databases (Steuer and Junker, 2009; Kanehisa and Goto, 2000).

Reaction network-based models have been published in the literature for variousorganisms and applications, and range from small to large, and from simple tohighly complex. Rejc et al. (2017) has provided an informative graph of theevolution of CHO models as a function of time, with the models categorized aseither constraint-based (steady-state) or dynamical (kinetic), and with the numberof reactions indicated via marker size (Figure 3 in their paper).

Accordingly, most metabolic networks published in the literature have beenof relatively small-scale (Zamorano et al., 2010; Ben Yahia et al., 2015; Rejcet al., 2017). An early CHO metabolic network from 1999 (Nyberg et al., 1999)included 33 reactions to represent the central carbon metabolism responsible for thegeneration of energy, biomass building blocks and major waste products. With time,the size and level of detail of CHO metabolic networks have gradually increased(Rejc et al., 2017); yet, many of subsequent networks have remained of similar sizeas the one by Nyberg et al. (Provost and Bastin, 2004; Provost et al., 2005; Nolanand Lee, 2011). Minimally, the glycolysis and lactate secretion, the glutaminolysisand ammonium secretion, and the TCA-cycle are included, as well as a simplifiedrepresentation for the synthesis of new cells to simulate the cell growth, as in e.g. thenetwork by Provost and Bastin (2004). Reactions from the amino acid metabolismmay also be included, as in for example the networks by Gao et al. (2007), and Niuet al. (2013).

Networks with a large number of reactions and complex connectivity are im-practical for certain modeling applications, and in particular for kinetic modelsassociated with lack of knowledge about kinetic mechanisms and parameters (Zengand Bi, 2006). When developing a model that simulates a small set of the mostrelevant process variables, completeness becomes a subordinate ideal. The goalis instead to obtain a calculable model within a reasonable time frame, that canbe identified using available data. Thereby, ideals such as simplicity and 1-Causalcould serve as guiding ideals.

Kinetic model development in particular tends to arrive at minimal represen-tations of the metabolism. Starting from a slightly more detailed model, a modelreduction can be achieved by lumping, meaning that several interconnected reactionsare combined into an overall reaction. The complexity of the model can be furtherreduced by assuming a net direction for each reversible reaction. Based on fluxanalyses, the reactions with low flux can even be removed from the model (p. 70),if considered unimportant for the intended application (Gao et al., 2007; Naderiet al., 2011; Niu et al., 2013). A need for more detailed reaction networks in kinetic

44

Page 63: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.3. Introduction to metabolic reaction networks

models has been recognized (Zamorano et al., 2010, 2013); but this will requirethe development of suitable methodology and more information about the kineticmechanisms and regulation within the cells.

Larger and more detailed models have been established for constraint-basedsteady-state modeling applications, which do not consider kinetics. The earlygenome-scale CHO networks were derived from mouse models (Selvarasu et al.,2012; Martínez et al., 2013). The reconstruction of an actual CHO cell metabolicmodel had to wait for the completion of CHO genome annotation data. In 2011,the CHO genome had been sequenced (Xu et al., 2011), and several years later, in2016, a first consensus genome-scale metabolic model of CHO cell metabolism waspresented (Hefzi et al., 2016; Harcum and Lee, 2016; Rejc et al., 2017).

3.3.3 Genome-scale models

A genome-scale model covers all the reactions of the organism currently known,and as new information becomes available it is incorporated into the model. Thereconstruction of genome-scale models could therefore be described as a practice ofGalilean idealization, driven by the completeness ideal. Even though the genome ofan organism can be sequenced, a full characterization of the organism’s metabolismhas no clear ending (Steuer and Junker, 2009); metabolism does not only dependon the genomic data, and is situation-dependent. The transcriptome, proteome andmetabolome in particular are bound to change with the environmental conditions(see p. 53). Nevertheless, a consensus reconstruction possesses a high degree ofgenerality, and can be re-used for many kinds of problems (Almquist et al., 2014).

Genome-scale modeling is well established for microorganisms, e.g. variousspecies of bacteria (Feist et al., 2007; Orth et al., 2011) and yeast (Lopes and Rocha,2017). E. coli in particular has been extensively modeled (Feist et al., 2007; Orthet al., 2011). The BiGG Models database organizes published genome-scale modelsin a standardized format (King et al., 2016). Out of the > 80 genome-scale modelsin the database, more than 50 are of E. coli, but only a small number of mammalianmodels are available and are limited to few species.

Four BiGG models are of the human species (Homo sapiens). Naturally, thereis great interest in genome-scale modeling for applications in human health anddisease, and for better understanding human metabolism (Cook and Nielsen, 2017).One model of mouse (Mus musculus), probably the most commonly used mam-malian research model, is available (Sigurdsson et al., 2010). The CHO consensusreconstruction iCHO (Hefzi et al., 2016) from 2016 is the recent addition to the

45

Page 64: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

database, and has been appointed a significant advance for the CHO cell biotechnol-ogy community (Harcum and Lee, 2016).

Hefzi et al. (2016) introduces a global CHO model, iCHO1766, made up of1,766 genes, 6,663 reactions, and 4,456 metabolites, together with several CHO cellline-specific models, including CHO-K1; each has ⇡ 1,200 genes, 4,600 reactions,and 2,800 metabolites. In terms of compartmentalization, nine compartments areaccounted for, namely: the cytosol and the extracellular space, the mitochondria,the nucleus, the endoplasmic reticulum, the Golgi complex, the lysosome, theperoxisome, and the mitochondrial inter-membrane space.

3.3.4 Metabolic networks in the present work

The metabolic networks in this thesis are listed in Table 3.4. The list displays thegradual increase in complexity, starting from the smaller networks in papers I andII (N30 and N1), and ending with a genome-scale network (NG) in paper IV. Thesame gradual increase is visualized in the bar chart in Figure 3.2.

The smaller networks N30 and N1 both encompass ⇡30 reactions, on par withsimple CHO networks in the literature (Rejc et al., 2017). The focus of these simplenetworks are on the major metabolic subsystems in CHO cells, i.e. the glycolysis,the TCA-cycle, and the amino acid metabolism and biomass synthesis.

Networks N2 (Paper II) and N126 (Paper III) were both derived from the detailednetwork of CHO cells by Zamorano et al. (2010), and encompass ⇡ 100 reactions(the map of N126 is found on p. 106). Compared to the smaller networks, additionalpathways are now represented, e.g. the PPP, the urea cycle, lipid metabolism, thenucleotide synthesis, the fatty acid synthesis, and the protein synthesis. Furthermore,N126 includes the synthesis of IgG, and the intracellular space is separated into amitochondrion and the cytosol.

Finally, the network in Paper IV encompasses ⇡994 reactions divided into 13metabolic subsystems and 65 di↵erent pathways. This network is a genome-scalemodel, and is a reduced version of the mouse-derived genome-scale CHO model by(Selvarasu et al., 2012), here referred to as NGS.

3.4 Introduction to metabolic flux analysis (MFA)Metabolic flux analysis (MFA) and related methods deal with the distribution ofintracellular flux in metabolic networks, and in particular changes in this distributionresulting from perturbations, e.g. in the environment, or in the genome (Altamiranoet al., 2013). The calculation of an unknown flux vector v, which defines the flux

46

Page 65: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.4. Introduction to metabolic flux analysis (MFA)

Table 3.4: Metabolic networks in the present work. N2 and N126 were based on (Zamoranoet al., 2010), and NG on (Selvarasu et al., 2012). NG is a reduced version of the originalnetwork by Selvarasu et al. (2012); the values within parentheses refer to the originalnetwork. PA: pathway analysis. Glycol.: Glycolysis. TCA: TCA-cycle. Met.: Metabolism.Synth.:Synthesis. Prot.: Protein. AAM: Amino acid metabolism.

Pape

r

Subj

ect

Code

Reac

tions

Reve

rsibl

e

Meta

bolit

esEx

trace

llular

Meta

bolic

subs

ystem

s

I Kineticmodel

N30 30 18 38 23 Glycol., TCA, AAM,biomass synth.

II PA N1 36 7 31 22 Glycol., TCA, AAM,biomass synth.

N2 100 29 96 24 Glycol., TCA, AAM,PPP, urea cycle, lipidmet., nucleotidesynth., fatty acidsynth., prot. synth.,biomass synth.

III Kineticmodel

N126 126 74 118 29 Glycol., TCA, AAM,PPP, urea cycle, lipidmet., nucleotidesynth., fatty acidsynth., prot. synth.,biomass synth., mAbsynth..

IV PA NG 994 467 700 61 65 pathways dividedinto 13 subsystems.

(NGS) (1,545) (679) (1,351) (218) (69 pathways dividedinto 13 subsystems.)

over each network reaction, provides a quantitative characterization of the cellphenotype based on extracellular metabolite concentration measurements.

47

Page 66: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

N30 N1 N2 N126 NG NGS

0

500

1,000

1,500

2,000

2,500

48 43 129 20

0

1,461

2,224

38 31 96 118

700

1,351

#

# Reactions # Metabolites

Figure 3.2: Comparison of network size in terms of: the number of reactions, referring tothe number of irreversible reactions when each reversible reaction has been split into tworeactions (see p. 57); and the number of metabolites. N30 is from Paper I. N1 and N2 arefrom Paper II. N126 is from Paper III. NG and NGS are from Paper IV, where NG is thereduced and adapted version of NGS.

3.4.1 Metabolic flux analysis

MFA is based on the application of material balances to the system under study,and is carried out under the assumption and approximation of a pseudo-steady state(Zupke and Stephanopoulos, 1995; Nyberg et al., 1999). It is assumed that theconcentrations of intracellular metabolites remain constant with time, and that theoverall net rate of the consumption/production of each intracellular metabolite iszero. Thereby, only the extracellular metabolites can accumulate or decrease inconcentration.

In cell cultures, this implies an ideal state with stable extracellular substrateuptake and product secretion rates. Constant environmental conditions can bring acell culture close to this state, e.g. as those achieved in the chemostat and perfusionmodes (Goudar et al., 2010). On the contrary, the batch or fed-batch modes arecharacterized by dynamic conditions. Yet, for such cultures, it is commonly assumedthat the cells will adapt quickly; the validity of the pseudo-steady state can then bejustified for distinct phases of the culture.

48

Page 67: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.4. Introduction to metabolic flux analysis (MFA)

Box 3.1: MFA

"Aext

Aint

#v ="

qext

0

#, v j � 0, j 2 Jirrev. (3.3)

The stoichiometry of the reactions in the metabolic network is defined in matrices Aext (rowsof extracellular metabolites) and Aint (rows of intracellular metabolites). Jirrev is the setof irreversible reactions. v is a solution to the fundamental equation of MFA (3.3) and isconsistent with the stoichiometry and irreversibility constraints of the metabolic network,the experimentally determined uptake/secretion rates in qext, and the pseudo steady-stateassumption (Quek et al., 2010).

In the equation of MFA, (3.3) in Box 3.1, the metabolic network is accountedfor via the stoichiometric matrices Aext and Aint, and the available extracellularmeasurements via the vector qext. While Aext, Aint and qext are known, v is anunknown flux vector to be calculated. Aintv = 0 follows from the pseudo-steady-state assumption.

MFA has been applied in several studies of CHO and hybridoma cell cultures.For example, studies have been conducted to investigate the e↵ects on metabolismfrom e.g. di↵erent carbon sources (Altamirano et al., 2006), di↵erent amino acid bal-ances (Xing et al., 2011), butyrate treatment (Carinhas et al., 2013), and temperatureshifts (Martínez et al., 2013).

An equation system with fewer equations than unknown flux rates is under-determined. This is often the case in MFA. The metabolic network can be quitedetailed, while experimental data, qext, are limited to a few metabolites. As a result,several flux vectors may satisfy (3.3). Measurements can also be associated withuncertainty, which increases the number of possible solutions.

Finding just one of many solutions provides limited information (Mahadevanand Schilling, 2003). As an alternative, intervals on each reaction flux can becalculated. Such approaches have has been demonstrated for CHO cell cultures,e.g. interval metabolic flux analysis (Zamorano et al., 2010), and the flux spectrumapproach (Llaneras and Picó, 2008). Flux variability analysis, FVA, is a similarapproach that will be introduced in Section 3.4.2.

More narrow flux intervals can be achieved via the introduction of additionalconstraints, as demonstrated by e.g. Zamorano et al. (2010); the inclusion ofrate data for additional metabolites, and an assumption regarding the amino acidcatabolism, led to more narrow intervals for several fluxes in the metabolic network.

The blue cone in Figure 3.3 illustrates a flux space of feasible solutions in three

49

Page 68: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

dimensions (the graph is a simplified description as metabolic networks usually havemore than three fluxes). A point in this graph represents a specific flux distribution,v = [v1 v2 v3]T . As point a lies outside the cone, that flux vector is not feasiblegiven the metabolic network. Point b lies within the blue cone, and is thus afeasible solution. The red cone, which lies within the blue cone, illustrates a moreconstrained flux space obtained after adding known extracellular uptake/secretionrates (qext) as constraints. As point b lies outside the red cone, this solution is nowinfeasible given the added data, but point c which lies within the red and the bluecone remains a feasible solution. Here, this graphical representation illustrates theflux space concept in MFA. For a more rigorous definition and representation ofrelevant flux cones and/or flux polyhedra, see Paper II of the present thesis, and thepaper on elementary flux vectors by Klamt et al. (2017).

3. Mathematical modeling of cells

rates (qext) as constraints. As point b lies outside the red cone, this solution is nowinfeasible given the added data, but point c which lies within the red and the bluecone remains a feasible solution. Here, this graphical representation illustrates theflux space concept in MFA. For a more rigorous definition and representation ofrelevant flux cones and/or flux polyhedra, see Paper II of the present thesis, and thepaper on elementary flux vectors by Klamt et al. (2017).

v1

v2

v3

a

c b

Figure 3.3: Graphical illustration of the flux space, a convex polyhedron cone in three-dimensions. A flux vector v is defined by the three fluxes v1, v2, and v3 and is graphicallyrepresented by a point in this three-dimensional space.

3.4.2 Flux balance analysis (FBA)

Flux balance analysis (FBA) is useful in situations when (3.3) is underdetermined.By defining an objective function (Feist and Palsson, 2010), an optimization problemcan be formulated (Equation 3.4 in Box 3.2). This problem can be solved e�cientlyusing linear programming, even for genome-scale reconstructions.

The objective function f is preferably based on a relevant biological assumptionabout the cells, e.g. the maximization of the growth rate. In that case, the solution vfound produces a maximized flux over the biomass synthesis reaction in the network.Additionally, all reaction fluxes in the network can be constrained by formulatingupper and lower bounds, [vmin, vmax]. Extracellular metabolite rate data (qext) can

50

Figure 3.3: Graphical and conceptual illustration of the flux space, a convex polyhedralcone in three-dimensions. A flux vector v is here defined by the three fluxes v1, v2, and v3,and is graphically represented by a point in this three-dimensional space.

3.4.2 Flux balance analysis (FBA)

Flux balance analysis (FBA) is useful in situations when (3.3) is underdetermined.By defining an objective function (Feist and Palsson, 2010), an optimization problemcan be formulated (Equation 3.4 in Box 3.2). This problem can be solved e�ciently

50

Page 69: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.4. Introduction to metabolic flux analysis (MFA)

Box 3.2: FBA and FVA

minimizev

fob j(v) = c1v1 + c2v2 + · · · + c jv j (3.4a)

subject to Aintv = 0 (3.4b)Aextv = qext (3.4c)qextnm,min Aextnmv qextnm,max (3.4d)vmin v vmax. (3.4e)

A flux vector v minimizes (or maximizes) an objective function fob j. fob j depends on one orseveral fluxes in v (3.4). The problem is constrained by: (i) the stoichiometric constraints (massbalances) provided by the metabolic network (3.4b); (ii) the constraints defined by experimentaldata 3.4c; and (iii) the capacity constraints that specify the allowed lower and upper boundson the fluxes over the reactions in the metabolic network (3.4e). Here are also constraints inthe form of upper and lower bounds on the uptake/secretion rates for unmeasured metabolites(3.4d). In FVA, each flux, v j, in the network is systematically minimized and maximized,potentially using the optimal objective value, fob j(v⇤), as a constraint.

using linear programming, even for genome-scale reconstructions.The objective function fob j is preferably based on a relevant biological assump-

tion about the cells, e.g. the maximization of the growth rate. In that case, thesolution v found produces a maximized flux over the biomass synthesis reaction inthe network. Additionally, all reaction fluxes in the network can be constrained byformulating upper and lower bounds, [vmin, vmax]. Extracellular metabolite rate data(qext) can also be included in the problem formulation. The solution v must thenmaximize the objective, while satisfying all of those additional constraints.

Within the graphic representation of the flux space, if the objective is to maxi-mize the flux v2, the solution is a point achieving a maximum value of v2, withinthe feasible solution space (Figure 3.3). Here, this point lies on the edge of the cone.But in general, the solution v is not necessarily unique; several di↵erent v may givean equally optimal solution to the problem.

Flux variability analysis (FVA) is variant of FBA, in which each reaction fluxover the entire network is systematically minimized and maximized (Selvarasu et al.,2012). This procedure yields an interval defined by an upper and a lower valueon each reaction flux. FVA can therefore be used to examine the flux space of theproblem. As in MFA, if the FBA problem is further constrained, more narrow fluxintervals can be achieved and the flux distribution more precisely characterized.

The use of FBA to model bacterial cells benefits from their ability to grow

51

Page 70: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

in simple media; bounds/constraints on the uptake rates for nutrients from themedium can be readily assumed (Feist and Palsson, 2010). Furthermore, it is areasonable assumption that a major evolutionary driving force for bacterial existenceis to replicate; this qualifies the use of growth as an objective function in the FBAproblem. Consequently, FBA has been extensively used in genome-scale modeling,e.g. to perform in silico knock-out experiments, examine strategies for metabolicengineering, and to study bacterial evolution (Alper et al., 2005; Pál et al., 2005).

The application of FBA to mammalian cells is considered less straightforward.Mammalian cells have a more complex metabolism (Section 2.3) and complexmedium requirements (Section 2.2.9). Nevertheless, FBA has been applied toanalyze the metabolism of CHO cells in several studies (Martínez et al., 2013; Souet al., 2015; Ivarsson et al., 2015; Huang et al., 2017), in some cases in combinationwith isotope labeling (Sengupta et al., 2011) or ’omics data (Selvarasu et al., 2012;Lewis et al., 2016).

3.4.3 Dynamic MFA and FBA

In dynamic flux analysis, time series of concentration measurements are transformedinto flux values (Lequeux et al., 2010). Flux analysis (either MFA or FBA) is carriedout for each time point in the series, leading to a sequence of solutions (Ahn andAntoniewicz, 2012). Di↵erent pseudo-steady states under dynamic conditions canthereby be accounted for (Almquist et al., 2014). But additional equations wouldneed to be formulated to connect those sequential solutions to each other, as isachieved in a kinetic model.

3.4.4 Isotope labeling techniques

Conventional MFA can be complemented with measurements obtained using isoptope-labeling techniques (Sengupta et al., 2011). The cells are fed substrates with labeledatoms (e.g. carbon-13), and these substrates are metabolized by the cells. Thepresence of the labeled atoms in metabolic intermediates and macromolecules isthen assessed by nuclear magnetic resonance (NMR) spectroscopy or mass spec-trometry (MS). Given a metabolic network, the flux vector is then calculated usinga combination of metabolite and isotopomer balancing.

The application of labeling techniques can provide more detailed informationon the distribution of intracellular fluxes than conventional MFA using extracellularmeasurements only. The techniques are however more experimentally and compu-tationally demanding. When comparing fluxes obtained using isotope techniques

52

Page 71: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.5. Introduction to pathway analysis

and conventional MFA for CHO cells in perfusion cultures, Goudar et al. (2010)found good agreement. Meanwhile, Ahn and Antoniewicz (2013) found discrepancybetween the estimated fluxes, specifically in the distribution of flux over the TCAcycle as its activity was overestimated when relying on conventional MFA.

3.4.5 ’Omics data

’Omics technologies refer to large-scale approaches that study the cell in a com-prehensive manner, rather than individual genes, proteins or metabolites. Popular’omics technologies include genomics, transcriptomics, proteomics, and metabolomics.’Omics data sets are generally large. Interpretation of those data, and the combina-tion and integration of data from multiple ’omics technologies can be achieved viaconstraint-based modeling (Huang et al., 2017), e.g. using MFA or FBA.

’Omics data complements conventional metabolite balancing. As realized ingenome-scale reconstructions, genomic data indicate genes present in the DNA ofthe organism. Transcriptomic and proteomic data indicate the genes actually tran-scribed and translated, respectively. Metabolomic data can identify and potentiallyquantify a range of metabolites, and serve as indicators for enzymatic activities.

The amount and applications of ’omics data from CHO cell cultures are increas-ing (Lewis et al., 2016; Huang et al., 2017). Nevertheless, as each CHO cell linehas a unique genome, transcriptome, proteome, and metabolome, it can be di�cultto compare ’omics data across di↵erent cell lines (Wurm, 2013).

3.5 Introduction to pathway analysisIn pathway analysis, a pathway is formed by combining a succession of reactions ina metabolic reaction network at pseudo-steady state; in the resulting overall reaction,the intracellular metabolites have been eliminated, leaving only the consumption ofextracellular substrates and consumption of extracellular products (Provost, 2006).The combination is stoichiometrically balanced, and the intracellular metabolitesparticipating in the pathway are consumed and produced at the same net rate; theydo neither decrease nor accumulate with time.

3.5.1 Pathways in a metabolic reaction network

In a set of pathways, L , the l:th pathway is defined by the vector,

pl = [p1, p2, p3, . . . pJ]T , (3.5)

53

Page 72: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

where each coe�cient p j indicates the involvement of reaction j in pathway l. pl isthus of the same length as the number of reactions in the network. The pathwaysare gathered in the matrix P = [p1, p2, p3, . . . pL], where column l refers to thel:th pathway, and row j refers to the j:th reaction in the metabolic network.

As an example, Figure 3.4 shows how the reactions of the glycolysis in asimple network can be combined into a pathway to connect extracellular glucose toextracellular lactate. The net reaction is the macroscopic reaction of the pathway.The conversion is stoichiometrically balanced; two moles of lactate are producedfor each mole of glucose consumed. Here, p1 = p2 = 1, and p3 = p4 = 2. Anyremaining reactions of the network are not used so p5, p6, . . . , pJ = 0.

!"#$

%$&'!$

(!&$

%$&)*+,-$

./*+,-$

00$1211/*#34+-!#56-76+-89:;<=$>>>>>>>>>>>>>>>>>>>>>>>>>>>>>>>>$1/*#3?#+/*@36$:$>>>>>>>>>>>>>>>>>>>>>>>>>>>>>>>>>$A$&)*+,-$>0$B$./*+,-$$??????????????????????????????????????????????????????$$C$$$$D$$$$$$$$EFG/@36$??????????????????????????????????????????????????????$A$$$$A$$$$A$&)*+,-$>0$A$&'!$$B$$$$A$$$$A$&'!$>0$B$(!&$$$$$($$$$B$$$$A$(!&$>0$A$!"#$$$$$H$$$$B$$$$A$!"#$>0$A$./*+,-$$??????????????????????????????????????????????????????$

!"

!"#$%&' ()' !"#$%&'( )*( #+( !,( )-($#.-)/.)%0.( -'#.1)+( &0+20+3( '"4-#.'&&5&#-($'4#6)&04'/(7&.'"4( 8'"4-#.'&&5&#-( 3&5.)/'9( #+:( ;#.'"4( 8'"4-#.'&&5&#-( &#.4#4'9( 6<( &0+'#-( .)$60+#1)+( )*(+'4=)-2(-'#.1)+/>(*)'!,(#+#&</0/(%-0+4()54(0+(,#4&#6>((+)'?&&5/4-#1)+()*(4@'($#.-)/.)%0.(-'#.1)+(0+(4@'($'4#6)&0.(+'4=)-2>(!(0/(4@'($#.-)/.)%0.(A5"(-#4'(0+(5+04(%$)&B.'&&C(:#<>(

!"#$%&'(")*+%,-.$/*0.123%4(,5*

6%,&,..3*

07*83).$(,91*

+:1;%$*.<*<%&'(;3%*2&,9-&5'*("#$%&'(")35*3&$)%=*>:$,9%$*"%,-.$/*#.123%4(,5*".,*#.12:,&?."&335*<%&'(;3%*

+:1

;%$*.

<*@6'*

A1&33%$*'%,*.<*@6'=*0.12:,&?."&335*<%&'(;3%*<.$*("#$%&'(")35*#.123%4*"%,-.$/'=*

!"#$%&!'#()&

!! "#$! "%&! '#(! )*+! ,-*! ./*! "%0! '#1! 2(%! '#&!

*+),&-

+(#.&

3! "444! 4!5!6! "644! 644!5!7! 8444! 4!5!9! 8644! 644!5!:! ;444! 4!5!<! ;644! 644!5!=! >444! 4!5!?! >644! 644!5!@! A444! 4!5!!34! '444! 4!5!33! .644! 644!5!36! ,444! 4!5!37! ,644! 644!5!39! )644! 644!5!3:! 24:4! :4!5!3<! 2B*#!

,-.&/"0#'-1'*2"0-'*3".',&456-/"72'"0'89:'8&//'8$/4$%&'#(D+'E,):'&(F%%-)#.@(*)-(,5&1%&'(,'4#6)&0.(G4#4'/(5/0+3(!"4-#.'&&5&#-(,'#/5-'$'+4/((

;"4&%54$%&'3"4&.'HIJ(K-)L)/4C(F>(M(N#/1+C(7>(O<+#$0.($'4#6)&0.($):'&&0+3(5+:'-(4@'(6#&#+.':(3-)=4@(.)+:01)+>(P)5-+#&()*(K-).'//(Q)+4-)&(IRC(SISESTU(8TVVR9>(HTJ(K-)L)/4C(F>C(N#/1+C(7>C(F3#4@)/C(G>W>(M(G.@+'0:'-C(X>EP>(,'4#6)&0.(O'/03+()*(,#.-)/.)%0.(,):'&/Y(F%%&0.#1)+(4)(QZD(.'&&/>(K-).'':0+3/()*(4@'(RR4@(?!!!(Q)+*'-'+.'()+(O'.0/0)+(#+:(Q)+4-)&C(T[UTET[U[(8TVV\9>(H]J(L)+(^#$%C(F>(M(G.@5/4'-C(G>(,'4#4))&(\>VY(*#/4(#+:(A'"06&'('&'$'+4#-<($):'/(#+#&</0/>(N0)0+*)-$#1./(TTC(I[]VEI[]I(8TVV_9>(

*3<0-=/&.#&2&047'`@0/( =)-2( 0/( /5%%)-4':( 6<( ^`Z( #+:( 4@'( G=':0/@( 7)L'-+$'+4#&(F3'+.<( *)-( ?++)L#1)+( G</4'$/( 8a?WWDaF9>( b'( 4@#+2( G'&'"0/C(G=0//>(Q5&45-'($':0#(0/(20+:&<(%-)L0:':(6<(?-L0+'(G.0'+1c.>('

>04%-.$3?-0'(Q@0+'/'( @#$/4'-( )L#-<( 8QZD9( .'&&/( #-'( =0:'&<( 5/':( =04@0+( 60)4'.@+)&)30.#&( 0+:5/4-<( *)-( &#-3'E/.#&'(%-):5.1)+()*(60)%@#-$#.'51.#&/>(QZD(.'&&/(4<%0.#&&<(.)+/5$'(&#-3'(#$)5+4/()*(3&5.)/'(#+:(3&54#$0+'(4)(3'+'-#4'('+'-3<C(60)$#//(#+:(4@'(%-):5.4()*(0+4'-'/4>(F(&#-3'(%#-4(0/(#&/)(.)+L'-4':(0+4)(4)"0.(6<E%-):5.4/(&#.4#4'( #+:( #$$)+0#( 4@#4( .#+( @#L'( +'3#1L'( 0$%#.4( )+( 4@'( .'&&/>( Q@#+30+3( %-).'//( %#-#$'4'-/C( '>3>( 4@'($':05$(.)$%)/01)+C(%Z()-(4'$%'-#45-'C(.#+(0+A5'+.'(4@'($'4#6)&0.(6'@#L0)-()*(4@'(.'&&/C(0:'#&&<(&'#:0+3(4)(#(&'//(=#/4'*5&($'4#6)&0/$>((,'4#6)&0.(A5"(#+#&</0/(#+:($):'&0+3(.#+(%-)L0:'(:''%'-(5+:'-/4#+:0+3()*($'4#6)&0.(/4#4'/(4-033'-':(5+:'-(:0d'-'+4( .5&1L#1)+( .)+:01)+/C( #/( ='&&( #/( %)4'+1#&( *)-( /0$5&#1)+( #+:( )%1$0e#1)+( )*( 60)%-).'//'/>( F($'4#6)&0.(+'4=)-2( .)+/0/4/()*( #( /'4()*($'4#6)&04'/( 8'"4-#.'&&5&#-( #+:( 0+4-#.'&&5&#-9( .)++'.4':( L0#( #( /'4()*(-'#.1)+/(8!"#$%&'@9>(`@'(+'4=)-2(.#+(6'(-'%-'/'+4':($#4@'$#1.#&&<(0+(4@'(*)-$()*(#(/4)0.@0)$'4-0.($#4-0">(`@0/( $#4-0"( :'c+'/( .)++'.1L04<( #+:( /4)0.@0)$'4-<( )*( 4@'( +'4=)-2>( ?+*)-$#1)+( )+( 4@'( -'L'-/060&04<( )*(/'&'.4':(-'#.1)+/(.#+(#&/)(6'(#::':>(,'4#6)&0.(A5"(0/(4@'(A)=()*($#//(4@-)53@(4@'/'(+'4=)-2(-'#.1)+/>(F//5$0+3(%/'5:)E/4'#:<(/4#4'(#+:(30L'+(#($'4#6)&0.(+'4=)-2(#+:(#(/'4()*('"4-#.'&&5&#-($'4#6)&0.(A5"(-#4'/C(4@'(0+4-#.'&&5&#-(A5"(-#4'/(.#+(6'(.#&.5&#4':(5/0+3(&0+'#-(#&3'6-#>((`@'( .)$%&'"04<( )*($#$$#&0#+( .'&&($'4#6)&0/$($#2'/($):'&( .)+/4-5.1)+( #( .@#&&'+30+3( 4#/2>( !&'$'+4#-<(A5"($):'/(8!,/9(.#+(6'(5/':(4)(*)-$($#.-)/.)%0.($):'&/(0+(=@0.@('#.@(!,(-'%-'/'+4/()+'(%#4@(4@-)53@(4@'(+'4=)-2( HICTJ>(`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b'( %-)%)/'( #($'4@):)&)3<( 6#/':( )+( !,( #+#&</0/( #+:(,0.@#'&0/E,'+4'+($):'&0+3('f5#1)+/C( #+:( 5/'( 4@0/( $'4@):)&)3<( 4)( 3'+'-#4'( #($):'&( *-)$( #( :#4#( /'4( )*( $5&1%&'($'4#6)&0.( /4#4'/()64#0+':(0+(QZD(.'&&(.5&45-'/(=04@(L#-<0+3(&'L'&/()*($':05$(#$0+)(#.0:/>(

8%-77A."73"B/"05%C' 8-//56-%5?-0D' *' E&=' */#-%"4F2' 4-'F50./&'/5%#&'50.'3-2B/&G'0&4=-%<7'(`@'($):'&(0/('"%'.4':(4)(.#%45-'($5&1%&'($'4#6)&0.(/4#4'/>(`@'(0+%54(+'4=)-2($5/4(.)L'-(#(-#+3'()*(%)//06&'(/.'+#-0)/( 4)( .#%45-'( 0$%)-4#+4( %@'+)$'+#( #L#0&#6&'( 0+( 4@'( :#4#>( `@'( ,!`F`DD;( #&3)-04@$( .)$%54'/( #&&(%)//06&'( %#4@=#</( 8!,/9( 4@-)53@( 4@'( 0+%54( +'4=)-2>( g)-( &#-3'( #+:( .)$%&'"( +'4=)-2/C( 4@'( +5$6'-( )*( !,/(6'.)$'/( %-)@0601L'&<( &#-3'>( `@0/( %)/'/( #( &0$04#1)+( 4)( 4@'($'4@):)&)3<( 4)( )+&<( 5/'( /0$%&0c':( +'4=)-2/( 0+(=@0.@(%#-4/()*(4@'($'4#6)&0/$(+'':':(4)($):'&(4@'(:#4#($#<(+)4(6'(-'%-'/'+4':>(D5-(3-)5%(@#/(0+01#4':(#(.-)//E:0/.0%&0+#-<( .)&&#6)-#1)+(=04@( 4@'( O0L0/0)+( )*( D%1$0e#1)+( #+:( G</4'$( `@')-<( 8^`Z9( =04@( 4@'( #0$( 4)(:'L'&)%(+'=($):'&0+3(4))&/>(b'(0$#30+':(#+(#&4'-+#1L'(#%%-)#.@(0+(=@0.@(4@'(3'+'-#1)+()*(!,/(4#2'/(4@'('"%'-0$'+4#&( :#4#( 0+4)( #..)5+4>( `@'( -'/5&4( 0/( 4@'( :'L'&)%$'+4( )*( 4@'( #( +'=( #&3)-04@$(6#/':( )+( #( .)+.'%4(.#&&':(.)&5$+(3'+'-#1)+(8Q79>(`@'(Q7(#&3)-04@$(0/(#6&'(4)(/4#-4(*-)$(&#-3'(#+:(.)$%&'"(+'4=)-2/>(?4(4@'+(5/'/(4@'( '"%'-0$'+4#&( :#4#( 4)( 04'-#1L'&<( 3'+'-#4'( %#4@=#</( 4@-)53@( 4@'($'4#6)&0/$( 4@#4( #-'($)/4( -'&'L#+4( 4)(:'/.-06'(4@'(:#4#>(G)(*#-C(4@'()54%54(/''$/(4)(6'(#(-'&#1L'&<(/$#&&(/'4()*(-'&'L#+4(!,/(#+:(4@'0-(.)--'/%)+:0+3(A5"(-#4'/(8!"#$%&'H(#+:(I9>(`=)($#h)-(#:L#+4#3'/()*(4@'(Q7(#&3)-04@$(#-'Y(

JGB&%"2&045/'*BB%-53F''K4%54&#C'D5-(4#-3'4(/</4'$(0/(4@'(.5&1L#1)+()*(#+(0+:5/4-0#&(QZD(.'&&(&0+'(%-):5.0+3(#($)+).&)+#&(#+16):<(%-):5.4(8$F69>(`)(3'+'-#4'(:#4#(*)-(4@'($):'&C(='(%'-45-6(4@'(/</4'$(6<(L#-<0+3(4@'(&'L'&/()*(/'&'.4':(#$0+)(#.0:/(0+( 4@'( .5&1L#1)+($':05$>(`@'/'(%'-45-6#1)+/(#-'('"%'.4':( 4)( 4-033'-( 4@'( .'&&/( 4)( /@0i(A5"( 0+($'4#6)&0.(%#4@=#</(#/(#(*5+.1)+()*(4@'0-('"4'-+#&('+L0-)+$'+4C(-'/5&1+3(0+(#(:#4#(/'4(-'%-'/'+1+3($5&1%&'($'4#6)&0.(/4#4'/(8!"#$%&'L9>((,&4F-.'`@'(QZD(.'&&( &0+'( 0/(#:#%4':(4)(3-)=(0+(/5/%'+/0)+(#+:(.5&1L#4':( 0+/0:'(/$#&&E/.#&'(G%0+(456'(60)-'#.4)-/(%&#.':(0+(#+(0+.56#4)->(`@'(.'&&/(#-'(.5&1L#4':(0+(4@'(%/'5:)E%'-*5/0)+($):'(8!"#$%&'M9>(?+(4@0/($):'C(4@'($':05$(0/(.)$%&'4'&<(-'+'=':()+(#(:#0&<(6#/0/j(4@'(.'&&/(#-'(@#-L'/4':(6<(.'+4-0*53#1)+C(/%'+4($':05$(0/(:0/.#-:':(#+:(4@'(.'&&(%'&&'4(0/(-'E/5/%'+:':(0+(*-'/@($':05$>(G#$%&'/(#-'(.)&&'.4':(6'*)-'(#+:(#i'-('#.@($':05$( '".@#+3'( #+:( #+#&<e':( *)-( .'&&( :'+/04<C( %'-.'+4#3'( )*( L0#6&'( .'&&/C( #+:( 4@'( .)+.'+4-#1)+( )*(0$%)-4#+4( '"4-#.'&&5&#-( $'4#6)&04'/( /5.@( #/( 3&5.)/'C( &#.4#4'C( #$$)+0#C( #$0+)( #.0:/( #+:( $F6>( `@'/'($'#/5-'$'+4/(#-'(5/':(4)(.#&.5&#4'(4@'(/%'.0c.($'4#6)&0.(A5"(-#4'/(.@#-#.4'-0e0+3('#.@($'4#6)&0.(/4#4'>(

!"#$%&'@>(G0$%&0c':(-'%-'/'+4#1)+()*(#($'4#6)&0.(+'4=)-2()*(QZD(.'&&($'4#6)&0/$(0+.&5:0+3(3&<.)&</0/C(`QFE.<.&'C(3&54#$0+)&</0/C(5-'#(.<.&'(#+:(#$0+)(#.0:($'4#6)&0/$>(`@'(/56/.-0%4('"4(0+:0.#4'/($'4#6)&04'/(0+(4@'('"4-#.'&&5&#-(/%#.'>(`@'($#h)-(.#-6)+(/)5-.'/(3&5.)/'(#+:(3&54#$0+'(#-'(/@)=+(0+(6&5'>(F$0+)(#.0:/(8'".'%4(3&54#$0+'9(#-'(/@)=+(0+(3-''+>(`@'($#h)-(6<E%-):5.4/(&#.4#4'C(#$$)+0#(#+:(.#-6)+(:0)"0:'(#-'(/@)=+(0+(-':>("##$%&'()*+,-.]K7C(]EK@)/%@)3&<.'-0.(#.0:j(F.Q)FC(F.'4<&(.)'+e<$'(Fj(#^7C(#&*#E^'4)3&54#-#4'j(F&#C(F&#+0+'j(F-3C(F-30+0+'j(F/+C(F/%#-#30+'j(F/%C(F/%#-4#4'j(Q04C(Q04-#4'j(Q</C(Q</4'0+'j(7_KC(7&5.)/'E_E%@)/%@#4'j(7&.C(7&5.)/'j(7&+C(7&54#$0+'j(7&5C(7&54#$#4'j(7&<C(7&<.0+'j(Z0/C(Z0/1:0+'j(?&'C( ?/)&'5.0+'j(;#.C(;#.4#4'j(;'5C(;'5.0+'j(;</C(;</0+'j(,#&C(,#&#4'j(,'4C(,'4@0)+0+'j(D"#&C(D"#&)#.'4#4'j(K@'C(K@'+<&#&#+0+'j(K-)C(K-)&0+'j(K<-C(K<-5L#4'j( G'-C( G'-0+'j( G5.C( G5..0+#4'j( G5.Q)FC( G5..0+<&( .)'+e<$'(Fj( `QF( .<.&'C( `-0.#-6)"<&0.( #.0:( .<.&'j( `@-C( `@-')+0+'j( `-%C( `-<%4)%@#+j( `<-C(`<-)/0+'j(a#&C(a#&0+'>(

8-04534'>01-%25?-0'(!E$#0&Y( (@#3-)4k24@>/'(b'6/04'Y( (===>.'4'3>/'B$*#(

95#%-4N'J)'5N''O-0P5/&P'9-745N'Q)'5N'*/"5#5N'*)'5N'RF50#N'S)'5N'*0.&%77-0N'*)'5N'Q53-67&0N'J)'T)'6N':..7.UV%N'9)'W)'3N'!-%7#%&0N'*)'3N'50.'8F-X&5$N'Y)'5'(

#(Q'&&(`'.@+)&)3<(7-)5%C(O0L0/0)+()*(?+:5/4-0#&(N0)4'.@+)&)3<C(^`Z (6(F54)$#1.(Q)+4-)&C((G.@))&()*(!&'.4-0.#&(!+30+''-0+3C(^`Z ( (.(O0L0/0)+()*(D%1$0e#1)+(#+:(G</4'$/(`@')-<C(O'%#-4$'+4()*(,#4@'$#1./C(^`Z(

Z%-B-7&.',-.&/"0#',&4F-.-/-#C((

!"#$%&'I)'!"%'.4':(%'-*)-$#+.'()*(4@'(Q7(#&3)-04@$(0+(.)$%#-0/)+(4)(,'4#4))&( \>V>(W5$6'-( )*( !,/( 8L'-1.#&( #"0/9( #3#0+/4( 0+.-'#/0+3(+'4=)-2(.)$%&'"04<(8@)-0e)+4#&(#"0/9>(

@'

L'

F( $'4#6)&0.( +'4=)-2( -'%-'/'+1+3( 0$%)-4#+4( $'4#6)&0.( %#4@=#</( 0/(.)+/4-5.4':(8!"#$%&'@9(#+:(-'%-'/'+4':(0+($#4-0"(*)-$#4>(?4(0/(#//5$':(4@#4(4@'(.'&&( )%'-#4'/( 5+:'-( %/'5:)E/4'#:<( /4#4'C( 0>'>( 4@#4( #&&( $'4#6)&0.( -#4'/( #-'(.)+/4#+4>((

M'

['

!&'$'+4#-<($):'/( 8!,/9(#-'(.)$%54':(5/0+3( 4@'(,'4#4))&(#&3)-04@$(H]J(#+:(0+4'-+#&($'4#6)&04'/('&0$0+#4':(6<(#&3'6-#0.()%'-#1)+>(!#.@(!,( 0/('f50L#&'+4(4)( #( $#.-)/.)%0.( -'#.1)+>( F( $#.-)/.)%0.( -'#.1)+( &0+2/( '"4-#.'&&5&#-($'4#6)&04'/( 6<( &0+'#-( .)$60+#1)+( )*( 4@'( -'#.1)+/( 0+( 4@'($'4#6)&0.( +'4=)-2(8!"#$%&'(9>((

`@'(A5"(4@-)53@('#.@($#.-)/.)%0.(-'#.1)+(0/($):'&':(6<((#(,0.@#'&0/E,'+4'+('f5#1)+(8!"#$%&'\9>(,#.-)/.)%0.(A5"(!(0/(4@'-'6<(#(*5+.1)+()*()+'()-(/'L'-#&(0+01#&(/56/4-#4'(.)+.'+4-#1)+/>(`@'(A5"(-#4'()*(#+('"4-#.'&&5&#-($'4#6)&04'(4@#4(0/()6/'-L':( 0+('"%'-0$'+4/(.#+(6'(.#&.5&#4':(6<( /4)0.@0)$'4-0.( /5$$#1)+()*(4@'(A5"(-#4'/(0+(4@)/'($#.-)/.)%0.(-'#.1)+/(4@#4(0+L)&L'/(4@0/($'4#6)&04'>(

`@'($):'&(%#-#$'4'-/( 8!/(0( #+:(129( #-'(cl':( 4)(:#4#(6<( &'#/4E/f5#-'/(:#4#(cm+3(8$0+0$0e#1)+()*(4@'('--)-(6'4=''+(:#4#E(#+:($):'&E:'-0L':(A5"(-#4'/9>((

!"#$%&' L)' F$0+)( #.0:( &'L'&( %'-45-6#1)+/( 4#6&'>( !#.@( -)=(-'%-'/'+4/( #( 4'/4( $':05$( 5/':( 0+( 4@'( QZD( .'&&( .5&45-'('"%'-0$'+4>( !#.@( .)&5$+( -'%-'/'+4/( #+( #$0+)( #.0:>( `@'( &'L'&/(#-'(30L'+(#/(%'-.'+4#3'/(0+(-'&#1)+(4)(4@'(&'L'&(0+(4@'(-'*'-'+.'($':05$(8Q4-&9>(((

!"#$%&'H)'g&)=(.@#-4(.)$%#-0/)+()*(4@'(,'4#4))&E6#/':(#%%-)#.@(#+:(4@'(Q7(F&3)-04@$>(*)',!`F`DD;(.)$%54'/(4@'(*5&&(/'4()*(!,/()*(4@'(0+%54( +'4=)-2>( g)-( .)$%&'"( +'4=)-2/C( 4@'( +5$6'-( )*( %#4@=#</(6'.)$'/(%-)@0601L'&<( &#-3'(#+:(4@'-'*)-'()+&<(/0$%&'(+'4=)-2/(.#+(6'(5/':>(+)'`@'(Q7(#&3)-04@$(5/'/(4@'(0+%54(+'4=)-2(4)(3'+'-#4'()+'(!,( #4( #( 1$'>( `@'( !,( 0/( 'L#&5#4':( #3#0+/4( 4@'( :#4#( 0+( #+( 04'-#1L'(*#/@0)+C(3'+'-#1+3(#(/56/'4()*(!,/($)/4(-'&'L#+4(4)(4@'(:#4#>(

;#-3'(#+:(.)$%&'"(0+%54(W'4=)-2(%)//06&'(!(533-$047'1-%'2-%&'6"-/-#"35/'B-77"6"/"?&7'

!"#$%&'\)',0.@#'&0/E,'+4'+($):'&0+3('f5#1)+>(!. 0/( 4@'($#.-)/.)%0.()-(!,(A5"( -#4'C( HGJ( 0/( 4@'(0+01#&( .)+.'+4-#1)+( )*( 4@'( '"4-#.'&&5&#-(/56/4-#4'C( #+:( !/(0( #+:( 1/( #-'( 20+'1.(%#-#$'4'-/>( `@'( 'f5#1)+( .#+( 6'( '"4'+:':( 4)(0+.&5:'( $5&1%&'( /56/4-#4'/( #+:( -'#.1)+(/4)0.@0)$'4-<(8+)4(/@)=+9>(

!"#$%&' M)' g&)=( .@#-4( 0&&5/4-#1)+( )*( %/'5:)E%'-*5/0)+( .5&1L#1)+( 0+(G%0+(456'(60)-'#.4)-/>(`@'(G%0+(456'/(#-'(%&#.':(0+(#+(0+.56#4)-(#4(]S( nQC( TVV( -%$(#+:(\(o(QDT>(D+(#(:#0&<( 6#/0/C( 4@'($':05$( 0+( '#.@(456'(0/(.)$%&'4'&<(-'+'=':(6<(.'+4-0*53#1)+>(G#$%&'/(#-'(.)&&'.4':(6'*)-'(#+:(#i'-($':05$('".@#+3'>(

G$#&&(/'4()*(!,/(#+:(.)--'/%)+:0+3(!,(A5"'/($)/4(-'&'L#+4(4)(O#4#(! &57"&%'4-'"04&%B%&4'6"-/-#"35//C'(

@' L'

!"#$%&'()"*+'+,

!"#$!"#$

%&'$

&()$

*+,$

*+,$

*+,$

-./)01233430)$56012$27/)01233430)$56012$

8$'9&$

%&'$

:1*;:$

0<'$

*-/$

=03$

+703$

>41*;:$>41$

?0127/$

8$'3127/$

@:A27/$

!"#27/$!"#$

*+,27/$

*+,$

*+,$

:5.27/$:5627/$*(527/$

'3(27/$

"-527/$B3227/$?2427/$

=2/27/$&);27/$&C227/$>2)27/$DC)27/$D)627/$D()27/$E0327/$

:3027/$

'3.27/$

!"#$

-./%,0*0"/, F)2027/$

:)G$"-5$&);$

B32$=2/$DC)$E03$

&C2$D()$

123,0*0"/,

!"*0)"*+'+,?24$&C2$D)6$?(5$B32$DC)$D()$

'34$ '3.$

:5.$:56$*(5$

'3($

"-5$B32$?24$

=2/$&);$&C2$>2)$DC)$D)6$D()$E03$

:30$

'3.$

:30$

'3($*(5$>2)$

:56$:5.$

&()27/$

>2)$:@-.;$01-H5$

I-;@05527/$

:)G27/$

'3427/$

?(527/$

:)G$

'34$

?(5$

!"#$%&'()*

+,-"*.$%/*

!"#$%&'()*+,#-

+&0,1/*

2-3#&)4*5(1$0/*

."/0(01"#2-&#-'%"03-4"2&(4-

."4,5"-%"4*&#&#)-0(1"%#*$*#$-

!"66-2"#0&$7-*#*670&0-

+&0,1/*

+&0,1/*

6)//7/)*6)//7/)*

!"#$%!"#$

!"#%%%&"%!%%%%%&$

!"#$%&'(

!"#$#%%&'()*'

+,'-&.%/0#12'

!"#$%&'(

)*#*(

+,-."#(%/(01.(

233'01.()*#*(

4"*.#5+6,*&".()*#*(789:;(

+,-."#(01(73,<".(

01(73,<".(

=:>&"*."?(>%@A3"<8#B(A%..8-3"(

2C(( DC((48@8#"?(.8E"(*:?(>%@A3"<8#B(

F,*38#*GH"I(JK*#(A*#K$*B.(?8?(#K"(>"33(,."L(

F,*:G#*GH"I(JK*#($"&"(#K"(M,<".(8:(#K"."(A*#K$*B.L(

233(A%..8-3"(A*#K$*B.(#K&%,;K(#K"(:"#$%&'(

F>( N>(1.#

2.#

3.#4.#

2#

2# 2#

1. 1 x Glcext => G6P2. 1 x G6P => 2 3PG3. 2 x 3PG => Pyr4. 2 x Pyr => Lacext

Glcext => 2 Lacext

Figure 3.4: A pathway linking extracellular glucose to extracellular lactate via the gly-colysis in a simple reaction network. w is the flux over the entire pathway of successivereactions. G6P, 3PG, and Pyr are intracellular metabolites (see p. ix). The macroscopicreaction is stoichiometrically balanced; two moles of lactate are produced for each mole ofglucose consumed.

The flux over the entire pathway of successive reactions is denoted by w. InFigure 3.4, when w is positive, glucose is taken up and converted into lactate via thethree intracellular metabolites G6P, 3PG, and Pyr. Intracellularly, no metabolitesaccumulate. The macroscopic reaction and its flux w can be applied to model thediminish of glucose and accumulation of lactate occurring in a cell culture. Acollection of macroscopic reactions that together link all the observable extracel-lular metabolites in the cell culture can form the basis for the development of a

54

Page 73: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.5. Introduction to pathway analysis

(grey-box) macroscopic kinetic model (Provost and Bastin, 2004; Provost et al.,2005; Zamorano et al., 2013). How to derive such models will be covered in Sec-tion 3.6 and 3.7 of this chapter. How to develop them for modeling the influence ofvaried amino acid availability in CHO cell cultures is the main focus in papers I andIII of this thesis.

3.5.2 The identification of relevant pathways

Given the complexity of most metabolic reaction networks, a relevant questionis how to identify its relevant and biologically meaningful pathways. Severalrelated concepts for systematic identification have been proposed, e.g. extremecurrents, ECs (Clarke, 2007), elementary flux modes, EFMs (Schuster and Hilgetag,1994), extreme pathways, EPs (Papin et al., 2002), and minimal generators, MGs(Urbanczik and Wagner, 2005). A thorough comparison of those concepts withexamples is available in the paper by Llaneras and Picó (2010). Among them, thereseems to be consensus that the EFMs are the most useful for the identification ofpathways that are biologically meaningful (Klamt and Stelling, 2003; Papin et al.,2004; Provost, 2006; Klamt et al., 2017).

All four concepts are related to the mathematical concept referred to as extremerays. Extreme rays are the edges of a convex polyhedral pointed cone (Llanerasand Picó, 2010), i.e. the edge vectors of the cone that generate the space of feasiblesolutions of v to the problem P · w = v, w � 0, see Figure 3.5. While P containsthe extreme rays and is referred to as a generating set, v can be obtained as anon-negative linear combination of P,

v = w1p1 + w2p2 + ... + wLpL, (3.6)

where wl � 0 for l = 1, 2, . . . , L. This decomposition is not necessarily unique,meaning that it is possible that v could be decomposed with di↵erent weight combi-nations (w) and onto di↵erent sets of pathways (P). The extreme rays in P constitutethe smallest generating set of the solution space, and are all the non-decomposablevectors in this space, meaning that each vector cannot be decomposed (brokendown) into simpler vectors.

Moving on to pathway analysis, the vectors in P represent pathways in a meta-bolic reaction network at steady state, thus Aintv = 0, and v is a feasible fluxdistribution over the individual reactions in the network. In the context of a macro-scopic cell culture model, the weights in w can be thought of as the pathway fluxes,like in Figure 3.4. The extreme rays of the cone in Figure 3.5 correspond to thepathways of a network in which all the reactions are irreversible (Llaneras and Picó,

55

Page 74: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells3. Mathematical modeling of cells

v1

v2

v3

Figure 3.5: Graphical illustration of the extreme rays of a convex polyhedron cone inthree-dimensions. A solution vector v = [v1, v2, v3] is graphically represented by a pointin this three-dimensional space. Feasible solutions lies within the cone. The extreme raysare the red edge vectors of the cone, and act as a basis for each feasible vector.

and reversible intracellular reactions. Consider, e.g., the metabolism of the NEAAsin CHO cells, of particular interest in the present work. The exchanges of thoseamino acids with the medium are reversible, and many of the intracellular reactionsinvolved in their metabolism are also reversible.

The definition and use of MGs, EFMs, or EPs, depend to a large extent onthe presence of reversibility in the network. The following three scenarios can beimagined: (i) all fluxes are irreversible; (ii) there are reversible reactions, but noreversible pathway; (iii) there are reversible reactions and reversible pathway(s) (seee.g. Figure 2 in the paper by Llaneras and Picó (2010)). In the simplest case, (i),there are only irreversible reactions; and there is a unique generating set of the fluxspace. This set is both the unique set of MGs, EPs and EFMs of the network. Butfor the development of the kinetic models considered in the present work, scenario(iii) is clearly the most relevant.

Extreme currents (ECs) (Clarke, 2007) were an early attempt to define pathwaysthrough a metabolic reaction network, that took reaction reversibility into account;each reversible reaction was split into two irreversible ones, running in the oppositedirections (Llaneras and Picó, 2010). This is referred to as moving from the originalvector space into an extended space of higher dimension, where all reactions are

56

Figure 3.5: Graphical and conceptual illustration of the extreme rays, of a convex polyhe-dron cone in three-dimensions. A flux vector v = [v1, v2, v3] is graphically representedby a point in this three-dimensional space. Feasible flux vectors lie within the cone. Theextreme rays are the red edge vectors of the cone, and act as a basis for each feasible vector.

2010). Each pathway is non-decomposable meaning that if one of the participatingreactions does not carry flux, the pathway cannot carry flux.

Reaction reversibility is a characteristic feature of metabolic reactions networks.In the general case, a network has both reversible transport reactions (exchanges)and reversible intracellular reactions. Consider, e.g., the metabolism of the NEAAsin CHO cells, of particular interest in the present work. The exchanges of thoseamino acids with the medium are reversible, and many of the intracellular reactionsinvolved in their metabolism are also reversible.

The definition and use of MGs, EFMs, or EPs, depend to a large extent onthe presence of reversibility in the network. The following three scenarios can beimagined: (i) all fluxes are irreversible; (ii) there are reversible reactions, but noreversible pathway; (iii) there are reversible reactions and reversible pathway(s) (seee.g. Figure 2 in the paper by Llaneras and Picó (2010)). In the simplest case, (i),there are only irreversible reactions; and there is a unique generating set of the fluxspace. This set is both the unique set of MGs, EPs and EFMs of the network. Butfor the development of the kinetic models considered in the present work, scenario(iii) is clearly the most relevant.

56

Page 75: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.5. Introduction to pathway analysis

Extreme currents, ECs) (Clarke, 2007), were an early attempt to define pathwaysthrough a metabolic reaction network, that took reaction reversibility into account;each reversible reaction was split into two irreversible ones, running in the oppositedirections (Llaneras and Picó, 2010). This is referred to as moving from the originalvector space into an extended space of higher dimension, where all reactions areirreversible. The ECs are obtained by finding the extreme rays to the pointed conein that extended space (Llaneras and Picó, 2010). When moving back to the originalspace, the reversible fluxes are merged together.

The elementary flux modes, EFMs (Schuster and Hilgetag, 1994), were a furtherextension of the concept of extreme rays, that accounted for reaction reversibility ina similar way as the ECs (Llaneras and Picó, 2010). The EFMs have the followingimportant properties:

• The set of EFMs for a network are all the non-decomposable pathway vectorsthat can generate the flux space. Because of this, the EFM set is exhaustive,and always unique for a particular network (Steuer and Junker, 2009), up to apositive scalar multiplication (Provost, 2006).

• The set of all EFMs is a generating set for the flux space, meaning that allfeasible flux distributions can be written as a non-negative linear combinationof the EFMs (Klamt et al., 2017).

• The EFMs are conformally non-decomposable vectors of the flux space(Klamt et al., 2017), because the elementary modes are defined with a "nocancelation rule", meaning that there must be no cancellation of reversiblefluxes. This rule is unique for the EFMs, and distinguishes them from otherpathway concepts (Llaneras and Picó, 2010). Importantly, after moving backto the original space, the EFMs contain the pathways necessary to generatethe flux space without cancelations.

• EFMs are not necessarily systemically independent, so an EFM could bea non-negative combination of other EFMs, as is illustrated in Figure 3.6(Llaneras and Picó, 2010; Papin et al., 2004).

Zanghellini et al. (2013) compared the tasks for EFM analysis to the task offinding the possible travel routes between two stations in a city subway system;EFM analysis can be used to identify the routes that pass a specific station, or thee↵ect of service interruptions on the set of possible routes. Similarly, EFM analysiscan be applied to answer questions about a metabolic network structure that are ofhigh relevance to e.g. metabolic engineering (Table 3.5).

57

Page 76: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

To answer the type of questions given in Table 3.5, two matrices are particularlyuseful. The first one is the matrix of the EFMs, denoted by E, in which each rowcorresponds to reaction j of the metabolic reaction network, and each column toEFM l; E specifies the reactions involved in each EFM. The second one is themacroscopic stoichiometric matrix, denoted by Amac, which is calculated via thefollowing matrix multiplication:

Amac = Aext · E (3.7)

In Amac, each row corresponds to an extracellular metabolite in Mext, and eachcolumn to a macroscopic reaction l; the l:th macroscopic reaction has been translatedfrom the l:th EFM in E. Amac specifies the extracellular substrates and products ofeach macroscopic reaction.

Table 3.5: Tasks for EFM analysis.

Task SolutionIdentify all the possible routes thatconnect two specific metabolites.

In Amac, find the columns that have a neg-ative element on the substrate row, and apositive element on the product row.

Identify the reactions that areessential to produce a certain prod-uct (Llaneras and Picó, 2010).

In Amac, find all the columns with a positiveelement on the product row. Among theassociated columns in E, find the rows ofthe reactions that have non-zero elementsin all the columns.

Compare pathways against eachother, e.g in terms of the number ofreactions used (Klamt and Stelling,2003).

Count the number of non-zero elements ineach vector el.

Identify the route between twometabolites with the highest yield(Klamt and Stelling, 2003; Llanerasand Picó, 2010).

In Amac, identify all the possible routes. Cal-culate the yield coe�cient for each routeby dividing the stoichiometric coe�cienton the product row with the one on the sub-strate row.

Examine the e↵ect of a "knock-out"of one or several reactions on thecapabilities of the network (Llanerasand Picó, 2010).

Identify the EFMs that involve one orseveral of the knock-out reactions by in-specting el for each route. Remove thosecolumns from E.

Each pathway in E is non-decomposable, meaning that all reactions of the

58

Page 77: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.5. Introduction to pathway analysis

pathway must carry flux for the pathway to carry flux. From a biological viewpoint,if the reaction carried out by a particular enzyme is not available (e.g. due to aknock-out of the corresponding gene or due to inhibition of the enzyme), pathwaysthat use that reaction are neither available. In the subway network example, routesthat pass stations in which service is currently interrupted will not be able to carrythe passenger to the terminal station via that route (Zanghellini et al., 2013).

The EPs will now be considered. The EFMs and EPs were compared againsteach other by several authors to clarify their definitions and possible applications(Klamt and Stelling, 2003; Papin et al., 2004; Llaneras and Picó, 2010). The EFMshave been found to be more useful in terms of analyzing the network properties(Klamt and Stelling, 2003; Klamt et al., 2017).

EPs are obtained in an extended space, but only intracellular fluxes are split intoforward and backward reactions; reversible transport reactions are kept as reversible.The EPs do not have the "no cancelation rule", and this could lead to cancelation ofreversible fluxes. Furthermore, to find the EPs means to identify all the pathwayvectors than can generate the flux space, and that are systemically independent inthe extended space; an EP can never be formed through a combination of other EPs.Unfortunately, this means that EPs can miss pathways present in the EFMs. As anexample, the EPs may miss optimal yield routes, or fail to identify the shortest andlongest route between a substrate and a product (Klamt and Stelling, 2003). Theissue is visualized in Figure 3.6, where the third pathway is an EFM, but not anEP, because it can be formed by combining EP 1 and EP 2. While EFM 3 wouldnot be found via an EP analysis, the pathway could still be of interest as a possibleroute between A and P. Finally, EPs need to be re-calculated if a reaction is removedfrom the network, or changed from reversible to irreversible (Klamt and Stelling,2003). With the set of EFMs, EFMs involving the associated reaction can simply beremoved from the set.

The last pathway concept, minimal generators (MGs), was devised to definea minimal number of systemically independent vectors that can generate the fluxspace. For scenarios (ii) and (iii) in which reversibility is considered, there could beEPs and/or EFMs that would not be included in the MGs. Because of this, MGs arenot very useful for assessing network properties as outlined in Table 3.5, but couldsu�ce when the goal is to find a small set that generates the flux space, and thatconnects all the extracellular substrates and products (Llaneras and Picó, 2010).

59

Page 78: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

EP 1

EFM 1

A

A C B

B

R1

R4 R5

R2

EP 2

EFM 2

B P

PB

R6

R3R2

EFM 3 A

A C B P

P

R1

R4 R5 R6

R3

Figure 3.6: Example of EFMs and EPs. The first two pathways are systemically indepen-dent and are both EFMs and EPs. The third pathway is only an EFM. It is systemicallydependent since it can be obtained by combining the two first pathways: EFM3 = EFM1 +EFM2 = EP1 + EP2. This figure is based on an illustration by Klamt and Stelling (2003).

3.5.3 EFM enumeration

EFM enumeration is to identify all the EFMs of a metabolic network. For complexmetabolic networks, finding any generating set is computationally challenging asa result of the combinatorial explosion of possible routes through the network. Inparticular, EFM enumeration becomes computationally prohibitive as the methodstrives to enumerate a very large number of pathways (Klamt and Stelling, 2002).

A good question is how many EFMs there are in a particular network. In a

60

Page 79: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.5. Introduction to pathway analysis

study by Provost et al. (2005), a simple network representing the growth phase ofthe CHO cell metabolism had 24 reaction fluxes, and was associated with just 11EFMs. Meanwhile, a model of E. coli with ⇡ 100 reactions was estimated to have⇡ 272 million EFMs (Jungreuthmayer and Zanghellini, 2012; Zanghellini et al.,2013). Clearly, the number of EFMs grows rapidly with increasing network sizeand complexity.

While metabolic reaction networks are often referred to by the number ofreactions, to estimate the number of EFMs in a particular network, structuralinformation must also be considered. Two equally-sized networks could havedi↵erent numbers of EFMs (Klamt and Stelling, 2002). Reversible reactions, andoccurrence of highly connected metabolites, e.g. some intermediates of the TCA-cycle (Steuer and Junker, 2009), are factors that tend to increase the number ofEFMs. This will become evident in Chapter 5 and in Paper I, in which one of thetasks is to enumerate the EFMs of a network that represents the flexible amino acidmetabolism of the CHO cells.

The standard approach for EFM enumeration is based on the double descriptionmethod (Fukuda and Prodon, 1996). The method generates an EFM candidate viapairwise combination of existing EFMs, and verifies that the EFM has not beenpreviously identified. The computation process requires high amounts of primarydata storage (Zanghellini et al., 2013). Efmtool (Terzer and Stelling, 2008) andMetatool (von Kamp and Schuster, 2006) are stand-alone tools based on the doubledescription method, and Metatool was applied in Paper I and II of this thesis.

When an enumeration of the EFMs is prohibitive, a subset of the EFMs maysu�ce. As an example, de Figueiredo et al. (2009) suggested an approach in whichthe analysis was limited to the k shortest EFMs. Kaleta et al. (2009) developeda concept referred to as elementary flux patterns to examine feasible pathwaysthrough subnetworks. Jungers et al. (2011) used the decomposition of a flux vectorestimated from experimental data to find a small and suitable subset of EFMs thatcould then be applied in macroscopic kinetic modeling (Zamorano et al., 2013).The idea of finding a relevant EFM subset is the focus in Paper II of this thesis.

3.5.4 EFMs-based MFA

EFMs-based MFA (Provost, 2006) combines the theory of MFA and EFMs totransform network fluxes v to pathway fluxes w, and provides a link between theexperimental data qext and w. Similar to conventional MFA, EFMs-based MFAcharacterizes the cell phenotype based on extracellular measurements, but from apathway-based point of view.

61

Page 80: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

The derivation of the EFMs-based problem starts from the fundamental MFAequation, here restated as (3.8),

"AextAint

#v ="

qext0

#, v j � 0, j 2 Jirrev. (3.8)

The flux vector v is decomposed on to the set of EFMs in E with weights w,

v = Ew. (3.9)

Equation (3.9) provides the important link between v and w, so that w can be linkedto qext. The external part of (3.8) is combined with (3.9), and (3.7),

qext

(3.8)#= Aextv

(3.9)#= AextEw

(3.7)#= Amacw, (3.10)

such that,qext = Amacw. (3.11)

Given qext, Aext, and E, an EFMs-based MFA problem is to find an estimate ofthe flux vector w that optimizes the quality of data-fitting,

minimizew

12

Xkqext � AextEwk22

subject to w � 0.(3.12)

In the original problem, the EFM set must be enumerated prior to the data-fitting,e.g. using the Metatool software (Provost and Bastin, 2004; Provost et al., 2005).

3.6 Kinetic modeling of cell populationsDynamic models that can simulate and predict the behavior of cells in di↵erentcultivation modes and under di↵erent culture conditions would be extremely usefulfor the biopharmaceutical industry. In the long-term, kinetic models will becomeincreasingly important for the improvement of industrial applications (Almquistet al., 2014). Over the years, a number of studies dedicated to the developmentof reaction network-based kinetic models of mammalian cell cultures have beenpublished (some of which are listed in Table 3.6), paving the way toward this goal.

This section begins with fundamental enzyme kinetics (Section 3.6.1), and ki-netics in cell population models (Section 3.6.2). The following sections focus specif-ically on the kinetics in macroscopic models derived from EFMs (Section 3.6.3),

62

Page 81: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.6. Kinetic modeling of cell populations

and on the related parameter estimation (Section 3.6.4) and network reduction(Section 3.6.5) in those types of models. Finally, the section ends with a discussionabout the scope of kinetic models, which is of interest when the goal is to capturediverse behavior and to identify optimal culture conditions (Section 3.6.6).

3.6.1 Enzyme kinetics

In a structured kinetic model, each reaction flux v j will be modeled by a kineticfunction,

v j = f j(c1, c2, ..., k1, k2, ...), (3.13)

where c1, c2, ... are metabolite concentrations and k1, k2, ... kinetic parameters. Asthe biochemical reactions within the cell are governed by the activity of enzymes,f j should logically be derived from the kinetics of the corresponding enzyme.

"The dynamics of metabolic networks are predominated by the activity ofenzymes - proteins that have evolved to catalyze specific biochemical trans-formations. The activity and specificity of all enzymes determines the specificpaths in which metabolites are broken down and utilized within a cell orcompartment."

Steuer and Junker (2009), p. 19.

The fundamental equation of enzyme kinetics is the Michaelis-Menten rateequation derived by Victor Henri (Henri, 1902) and evaluated by Leonor Michaelisand Maud Menten (Michaelis and Menten, 1913),

v = vmaxS

S + Ks, (3.14)

where v is the rate of the reaction, S is the concentration of a substrate, vmax isthe maximum rate of the reaction, and Ks is the Michaelis constant which is theconcentration of S at which the reaction proceeds at half of the maximum rate. Aplot of this rate equation is shown in Figure 3.7.

63

Page 82: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

Tabl

e3.

6:Ex

ampl

esof

publ

icat

ions

focu

sed

onki

netic

mod

els

ofC

HO

cells

and

othe

rmam

mal

ian

cell

type

s.r:

reac

tions

.m:

met

abol

ites.

MM

:Mic

hael

is-M

ente

n.

Cell

line

Mod

eNe

twor

kM

acro

-reac

tions

Kine

tics

Refe

renc

e

Stru

ctur

edm

odel

sC

HO

Fed-

batc

h24

m,3

4r-

Con

veni

ence

rate

law

s(N

olan

and

Lee,

2011

)C

HO

Bat

ch30

rM

onod/M

M(G

horb

ania

ghda

met

al.,

2013

)C

HO

Bat

ch35

r-

Mon

od/M

M(G

horb

ania

ghda

met

al.,

2014

)G

S-C

HO

Bat

ch/fe

d-ba

tch

24r

-M

M(C

hen

etal

.,20

14)

CH

O-D

XB

11B

atch/fe

d-ba

tch

34r

-M

onod/M

M(R

obita

ille

etal

.,20

15)

Rea

ctio

nne

twor

k-ba

sed

mac

rosc

opic

mod

els

CH

OB

atch

20m

,18r

7EF

Msd

Mon

od/M

M(P

rovo

stan

dB

astin

,200

4)C

HO

Bat

ch25

r3-

11EF

Msd

Mon

od/M

M(P

rovo

stet

al.,

2005

)H

ybrid

oma

Bat

ch32

r9

EFM

saM

onod/M

M(G

aoet

al.,

2007

)H

ybrid

oma

Bat

ch/

Fed-

batc

h30

m,3

2r10

EFM

saM

onod/M

M(D

orka

etal

.,20

09)

CH

OB

atch/

Fed-

batc

h30

m,3

4r8

EFM

saM

onod/M

M(N

ader

ieta

l.,20

11)

CH

OB

atch

72m

,100

r19

EFM

sbM

onod/M

M(Z

amor

ano

etal

.,20

13)

Hyb

ridom

a/G

S-C

HO

Perf

usio

n/Fe

d-ba

tch

35r

9EF

Msa,

dN

oki

netic

s(N

iuet

al.,

2013

)

CH

O-K

1SV

Pseu

do-p

erfu

sion

38m

,30r

379

EFM

sdM

onod/M

MPa

perI

CH

O-K

1SV

Pseu

do-p

erfu

sion

118m

,126

r12

5EF

Msc

Mon

od/M

MPa

perI

IIa

EFM

sde

rived

afte

rred

uctio

nof

the

netw

ork

base

don

apr

iori

MFA

.b

Asu

bset

ofth

eEF

Ms

deriv

edus

ing

ade

com

posi

tion

ofa

flux

vect

or(J

unge

rset

al.,

2011

).

cA

subs

etof

the

EFM

sde

rived

usin

gth

eco

lum

n-ge

nera

tion

tech

niqu

e(P

aper

III)

.d

EFM

enum

erat

ion

with

Met

atoo

l(vo

nK

amp

and

Schu

ster

,200

6).

64

Page 83: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.6. Kinetic modeling of cell populations

S

vvmax

vmax2

Ks

v = vmaxS

S+Ks

0 1 2 30

1

2

3

4

Figure 3.7: Michaelis-Menten–type equation where vmax = 4 and Ks = 0.5.

The fundamental Michaelis-Menten rate equation describes a substrate satu-ration e↵ect imposed by a single substrate on the reaction rate v; the function ishyperbolic and approaches vmax for increasingly higher values of S . At zero sub-strate concentration, v = 0; logically, if the substrate is not present, the reactioncannot proceed.

The Michaelis-Menten equation is an approximative and simplified descriptionof the kinetics of most enzymes. The derivation of (3.14) rests on numerousassumptions (Steuer and Junker, 2009). For example, it is assumed that: thesubstrate binds reversibly to the enzyme, forms an enzyme-substrate complex, andthat the formed product then dissociates irreversibly; the binding of the substrateis much faster than the slow dissociation of the product; the complex immediatelyreaches steady-state as the reaction begins, and rapidly adjust to slow variations inthe substrate concentration; and that the concentration of enzyme-bound substrate isnegligible compared to the total substrate concentration (Steuer and Junker, 2009).

When a product is present at high concentration, the reversible version ofthe reaction should be derived, incorporating product inhibition e↵ects (Steuerand Junker, 2009). Furthermore, most enzymatic reactions involve two or moresubstrates, and substrates and products can bind and dissociate in di↵erent ways.There can also be allosteric regulation, in which a molecule binds to the enzymeto increase or decrease its activity. Consequently, there are many possible rate-equations, many of them are tedious to derive, and for which parameter estimationthrough experiments is di�cult (Steuer and Junker, 2009).

65

Page 84: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

3.6.2 Kinetics in cell population models

It has been emphasized that for the kinetic modeling of cell populations, a globalsystem perspective is necessary (Almquist et al., 2014). Even though kineticparameters for enzymes can be found in databases, e.g. BRENDA (Barthelmeset al., 2007), those rate laws were usually derived in vitro and under di↵erentconditions than those occurring in a living cell (Chen et al., 2014; Almquist et al.,2014). The allosteric regulation within the network is an important aspect in thiscontext, but is di�cult to characterize (Steuer and Junker, 2009).

Instead of trying to elucidate detailed kinetics, cell population models tend tobe developed from approximative and generalized kinetic equations, and judgedby their ability to capture the global behavior of the modeled system (Steuer andJunker, 2009). This solution suits automation (Almquist et al., 2014). Examples ofgeneralized kinetic functions are power-laws and linearizable logarithmic equations(Steuer and Junker, 2009; Almquist et al., 2014; Ben Yahia et al., 2015), andMichaelis-Menten–type equations (Table 3.6).

Convenience kinetics was developed to model kinetics in metabolic reactionnetworks (Liebermeister and Klipp, 2006a,b). In this approach, each reaction ofa metabolic network can be systematically gone through to determine a rate equa-tion of Michaelis-Menten–type, accounting for potential saturation, activation andinhibition e↵ects on the reaction flux. Parameters that ensure that thermodynamicconstraints are met can also be included. The approach was applied by Nolan andLee (2011) to model CHO cell cultures; there the general equations were manuallyadjusted based on an experimental data base, i.e. on the quality of the data-fitting,and complemented by the introduction of regulatory factors.

The Monod-equation, named after Jacques Monod, models the experimentallyobserved growth rate of a cell population of a microorganism as a function ofa limiting nutrient concentration (Monod, 1949). The Monod equation does notrequire any structural information available in metabolic reaction networks, and canbe derived from data. In unstructured animal cell population models, Monod–typeequations have been frequently applied to describe the experimentally observedmacroscopic behavior of the cultured cells (Zeng and Bi, 2006; Ben Yahia et al.,2015). The cell growth rate is a function of one or several limiting substrateconcentrations in the medium, e.g. glucose and/or glutamine. The kinetic parametersare the maximum cell-specific growth rate, µmax, and a substrate half-saturationconstant, KS , for each limiting substrate, such that the general Monod-equation is:

µ = µmax

Y

i

S i

S i + KS i

= µmax ·S 1

S i + KS 1

· S 2

S 2 + KS 2

. . .S i

S i + KS i

. (3.15)

66

Page 85: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.6. Kinetic modeling of cell populations

For a single substrate, (3.15) resembles (3.14), the fundamental Michaelis-Mentenequation for enzyme kinetics. Inhibition from secreted by-products can also beaccounted for; here follows an example from an unstructured model of hybridomacells (Jang and Barford, 2000; Ben Yahia et al., 2015) that accounted for saturatione↵ects from limiting nutrients glucose and glutamine, and inhibitory e↵ects fromby-products lactate and ammonium:

µ = µmax ·Glc

Glc + KGlc· Gln

Gln + KGln· kLac

Lac + KLac· kAmm

Amm + KAmm. (3.16)

Rates other than µ, e.g. qGlc, and qGln, can be calculated from known yield co-e�cients, e.g. YBiomass/Glc, and YBiomass/Gln. Those rates are then proportional tothe growth rate. An alternative is to derive additional Monod–type equations (BenYahia et al., 2015).

3.6.3 Macroscopic kinetic models derived from EFMs

In macroscopic kinetic models derived from EFMs, a kinetic function for eachpathway l is defined, and the full set of functions are collected in F such that,

qext = Aextv = AextEw = AextEF(c1, c2, ..., k1, k2, ...)wmax (3.17)

Each macroscopic flux has thus been replaced by a kinetic function. This approachhas been applied in several papers (Provost and Bastin, 2004; Provost et al., 2005;Gao et al., 2007; Dorka et al., 2009; Naderi et al., 2011; Zamorano et al., 2013), aswell as in papers I and III of the present thesis.

When formulating a macroscopic rate equation, it could be reasoned that asthe individual reactions making up a pathway are governed by enzymes, enzymekinetics could be applied. But even if rate laws could be derived for each of theparticipating enzymes, it is not a straightforward task to combine those rate lawsinto a rate law for the entire pathway. Given a rate-limiting step of the pathway,kinetics could perhaps be derived from the kinetics of the governing enzyme. Itappears however that the control of flux is shared among multiple steps rather thanby single enzymes (Moreno-Sánches et al., 2008); pathways do not proceed inisolation, but are interconnected with other pathways.

But as Monod–type equations can model the global behavior of cells, a macro-scopic model built on general Monod– or similar Michaelis-Menten–type equationsappears reasonable, at least as a starting point in model construction (Provost et al.,2005; Zamorano et al., 2013). As seen in Table 3.6, the kinetics in the macroscopic

67

Page 86: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

models are all based on generalized multiplicative Monod- or Michaelis-Menten–type kinetics. The general equation of this type, also adopted in the present work,can be stated as follows,

wl = wmax,l

Y

i

S ext,i

S ext,i + KS ext,i

, (3.18)

were wl is the macroscopic flux rate, wmax,l is the maximum macroscopic fluxrate, S ext,i is the initial concentration of the extracellular substrate i and KS ext,i isits associated half-saturation constant. Plots of this equation at several di↵erentvalues for KS ext are shown in Figure 3.8, as is the corresponding plots for productinhibition.

3.6.4 Parameter estimation and the balanced-growth assumption

An e�cient estimation of multiple parameters in several simultaneous non-linearequations can be a challenge, especially for large equation systems, see e.g. (BenYahia et al., 2015) for approaches applied in macroscopic modeling. To simplifythe estimation of the kinetic parameters in (3.18), the non-linear problem can betransformed into a linear problem. Values for each half-saturation parameter KS ext,i

is specified beforehand and fixed. In the linear problem, the vector wmax is estimatedfrom the experimental data (Provost et al., 2005; Gao et al., 2007; Zamorano et al.,2013).

The simplification is justified by a balanced-growth assumption; as substratesare available in excess, it is assumed that the macroscopic reactions occur at theirmaximum rates (Gao et al., 2007; Zamorano et al., 2013). This assumption isrealized in a model by setting each KS ext,i to a low value relative to the initialconcentration of the substrate (Provost et al., 2005; Gao et al., 2007; Zamorano et al.,2013). The principle is illustrated in Figure 3.8 for a case in which KS ext = 0.001.In that case, w ⇡ wmax, unless S ext is very small or zero. This principle will berevisited in Chapter 5, when examining the application of generalized kinetics in amacroscopic kinetic model that is supposed to model the influence of varied aminoacid availability on the macroscopic behavior of sixteen parallel CHO cell cultures.

3.6.5 Network reduction in preparation for kinetic modeling

Most of the metabolic networks applied in the reaction network-based macroscopickinetic models listed in Table 3.6 were defined by less than 40 reactions. The

68

Page 87: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.6. Kinetic modeling of cell populations

S ext

w

Ks = 10

K = 2

Ks = 1Ks = 0.5Ks = 0.1

Ks = 0.001

Ks = 1000

0 1 2 30

1

2

3

4

S ext

wKs = 0.001

Ks = 1000

0 1 2 30

1

2

3

4

Pext

wKp = 1000

Kp = 10

Kp = 5

Kp = 1Kp = 0.5Kp = 0.1

Kp = 0.0010 1 2 3

0

1

2

3

4

Pext

wKp = 1000

Kp = 0.0010 1 2 3

0

1

2

3

4

Figure 3.8: A plot of the equations w = wmaxS ext

(S ext+Ks)(top) and w = wmax

Kp

(Pext+Kp) (bottom)for di↵erent values of Ks and Kp, and where wmax = 4. Here, S ext is the initial concentrationof a single extracellular substrate, Pext is the initial concentration of a single extracellularproduct, and w is the macroscopic flux rate. The dashed line indicates an initial concentrationof 0.2 mM.

69

Page 88: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

network in the EFMs-based model developed by Zamorano et al. (2013) is anexception, with 100 reactions.

In a priori MFA, prior to the identification of pathways, a flux vector v iscalculated to determine a subnetwork of the most significant reactions; reactionswith small fluxes are removed. Gao et al. (2007), Naderi et al. (2011), and Niu et al.(2013) each used the approach to reduce their initial networks in preparation forkinetic model development. Starting from metabolic networks of approximately 30reactions, most of them irreversible, after network reduction their resulting kineticmodels were developed from eight or nine macro-reactions deemed to be the mostsignificant (Gao et al., 2007; Naderi et al., 2011; Niu et al., 2013).

Now, the results of the a priori MFA in those studies can be looked at in termsof the amino acid metabolism, which is central to the present thesis. The aminoacid metabolism was not well represented in those models. Gao et al. (2007) endedup removing the catabolism and interconversions of a majority of the amino acids.For Naderi et al. (2011), only five significant amino acids remained in the reducednetwork. Niu et al. (2013) assumed that aside from glutamine, alanine and glutamate,all other amino acids were used exclusively for protein synthesis.

In the model by Zamorano et al. (2013), a reduction of the complex 100 reactionnetwork was achieved by applying the algorithm developed by Jungers et al. (2011)to identify an EFM subset (Section 3.5.3, p. 61). A major advantage of this approachwas that the EFMs could be obtained directly from the detailed network; the networkdid not have to be reduced using e.g. a priori MFA. Keeping detailed reactions ofthe catabolism and interconversions of the amino acids, the resulting kinetic modelby Zamorano et al. (2013) was able to simulate a CHO cell batch culture and, i.e.,the evolution of glucose, lactate, ammonium and 17 amino acid concentrations withtime.

3.6.6 Modeling diverse conditions

The scope of a model refers to the situations the model is applicable to, and is clearlyof high relevance for anyone that intends to use it. A model might be applicable toa certain mode of cultivation, to a specific phase of a culture, cell line, or mediumcomposition, etc. This is important to bare in mind when examining models andproposed modeling methodologies. Below follows examples from the literaturefocused on the kinetic modeling of diverse conditions and changing behavior of CHOcells, e.g. encountered during dynamic cultivation modes, from applying stimulatorychemicals, stemming from clonal variation, or di↵erent medium compositions.

70

Page 89: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.6. Kinetic modeling of cell populations

The simulation of batch or fed-batch cell cultures is of high relevance to the bio-pharmaceutical industry (Zhang, 2010); naturally, the majority of the kinetic modelsin Table 3.6 were developed from batch- or fed-batch culture data. Throughout suchcultures, the environment of the cells is changing with time; consequently, the cellspass through di↵erent phases as described in Section 2.2.5 (p. 13). A challengeis how to generate a kinetic model under such dynamic conditions (Provost et al.,2005; Gao et al., 2007; Zamorano et al., 2013; Robitaille et al., 2015).

One strategy is to generate several consecutive and phase-specific models. (Gaoet al., 2007) divided a batch culture into two phases corresponding to a growthand post-growth phase, and developed one model for each phase. The phase-specific models can also be combined by linear switching functions, for which thetime points at which the transitions occur are based on the actual data. Provostand Bastin (2004) considered only the phase of exponential growth in their initialmodel, but the stationary and death phases were accounted for in a second modelby switching functions, combining three phase-specific models to simulate theconcentration of cells and metabolites throughout the entire batch culture (Provostet al., 2005). Switching functions were again applied by Zamorano et al. (2013) tomodel the phases of a CHO cell batch culture, but now using the EFMs obtainedfrom the more detailed 100 reaction network.

Sodium butyrate is a chemical whose addition to cell cultures can improve therecombinant protein production (McMurray-Beaulieu et al., 2009). Ghorbaniagh-dam et al. (2013) studied the e↵ect of sodium butyrate stimulation in CHO cellcultures by developing kinetic models representing the cell metabolism with andwithout butyrate stimulation, respectively. Each condition was represented by aseparate model, to provide information about its metabolic pattern; those modelscould then be compared.

Metabolic e�ciency is of interest when selecting between di↵erent clonesfor biopharmaceutical production. This scenario was examined in another studyby Ghorbaniaghdam et al. (2014). Di↵erent CHO cell clones with and withoutinduction by cumate were investigated, and could be compared with the help ofclone-specific kinetic models (Ghorbaniaghdam et al., 2014; Robitaille et al., 2015).

The strategy of modeling distinct phases of batch cultures was further examinedby Robitaille et al. (2015), noting that the transitions between the phases happensdue to biological mechanisms and intracellular changes. Based on the model byGhorbaniaghdam et al. (2013), the authors generated a new structured model ofmAb-producing CHO-DXB11 cells in the batch/fed-batch mode. With access tointracellular metabolite data, the model was able to at least simulate the exponentialand early plateau-phase of the culture using a single set of parameters. Robitaille

71

Page 90: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

et al. (2015) tried simulations in two di↵erent culture media, and concluded thatwhile their model was likely cell line-specific, the approach did show potential forsimulating the behavior of a specific cell line in di↵erent media.

3.7 Derivation of a macroscopic kinetic modelIn this section, a macroscopic kinetic model is derived in eight steps by combiningthe concepts introduced in the previous sections. To help to visualize the procedure,a flow chart is presented in Figure 3.9.

Step 1: Perform experiment The model target is studied in cell cultures togenerate experimental data. The approach relies on an access to a typical cellculture laboratory, including the possibility to measure the viable cell number, theconcentrations of selected metabolites, and the concentration of the product atconsecutive time points. The corresponding rates of growth, µ, product secretion,qmAb, and metabolite uptake and secretion, qext, are calculated using equations (2.1),(2.3), and (2.4) in Chapter 2. The rates are collected in the vector,

qext = [qext,1 qext,2 qext,3 . . . , qext,Mext ]T , (3.19)

Here, Biomassext and mAbext belong to Mext and are extracellular products.

Step 2: Define metabolic reaction network The metabolism of the cell line isrepresented in a metabolic reaction network of the type introduced in Section 3.3.The network is specified by Aint, Aext, and virrev.

Step 3: Enumerate the EFMs The pathways accessible to the cells are identifiedvia systematic enumeration of the EFMs of the metabolic network. The EFMsprovide links between the extracellular substrate and product metabolites in Mext.The EFM enumeration is carried out using the Metatool software (Provost andBastin, 2004; Provost et al., 2005; von Kamp and Schuster, 2006) to which Aint andvirrev are given as inputs. Metatool provides the EFM-matrix E, and an irreversibilityvector eirrev to indicate EFMs reversible in the original space.

Step 4: Translate into macroscopic reactions Revisiting (3.7), the EFMs aretranslated into macroscopic reactions, Amac = Aext · E. In Amac, each row corre-sponds to an extracellular metabolite in Mext, and each column to macroscopicreaction; the l:th macroscopic reaction has been translated from the l:th EFM in E.

72

Page 91: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3.7. Derivation of a macroscopic kinetic model

1Perform

experiment

2Specify metabolicreaction network

3Enumerate EFMs

4Translate intomacroscopic

reactions

5State the EFMs-

based MFA problem

6Introduce

kinetic functions

7Estimate the

parameters andsimulate rates

8Evaluate

qext

qext

c0

K

Aint, virrev

E, eirrev

Amac

w

fl

wmax, qsim

Aext

Figure 3.9: Workflow for a derivation of a reaction network-based macroscopic kineticmodel, where qsim refers to the simulated rates.

73

Page 92: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

3. Mathematical modeling of cells

Step 5: State the EFMs-based MFA problem Revisiting (3.11), the EFMs-based MFA problem is stated, qext = Amacw, where w contains the macroscopicflux rates.

Step 6: Introduce kinetic functions Each macroscopic flux rate wl is modeled bya kinetic function based on a generalized Michaelis-Menten–type kinetic equation2,

wl = wmax,l · fl = wmax,l

Y

i2Mext,s,l

c0,i

c0,i + Ks,i,l= wmax,l

Y

i2Mext,s,l

c0,iKs,i,l

c0,iKs,i,l+ 1

(3.20)

where c0,i is the initial concentration of metabolite i, Ks,i,l is the half-saturationconstant of metabolite i in macroscopic reaction l, and Mext,s,l is the set of substratemetabolites in macroscopic reaction l.

Step 7: Parameter estimation The parameter estimation is carried out by solvinga least-squares-type minimization problem, e.g.,

minimizewmax

12

DX

k=1

���qext,k � AmacF(c0,k,K)wmax���2

2

subject to wmax � 0,

(3.21)

See Section 2.2 in Paper III for details about (3.21). Each Ks,i,l is fixed to a guessedvalue. Given an assumption of balanced growth (Provost and Bastin, 2004; Provostet al., 2005; Zamorano et al., 2013), Ks,i,l can be set su�ciently small such thatwl ⇡ wmax,l unless S s,i,l is very small relative to the initial concentration of thecomponent in the medium (see Figure 3.8, p. 69).

Step 8: Evaluation The rates simulated by the model, qsim, are compared to thedata qext (direct validation).

2 The rate-equation is rewritten to avoid division by a small number, something which couldhappen when Ks is small and c0 is small or equal to zero.

74

Page 93: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Chapter 4

Experimental data: Influence ofvaried amino acid availability inCHO cell cultures

This chapter presents the experiment carried out to generate data for the modeldevelopment in Chapters 5-8.

4.1 Aim and overviewRevisiting the aim of this thesis (p. 5), the methodology developed should be ableto generate one single macroscopic kinetic model with the ability to capture diversemetabolic behavior of the CHO-K1SV cell line in response to variations in the aminoacid availability. The reference medium, cultivation mode and type of bioreactorhave already been specified, and the workflow is illustrated in Figure 4.1 for thegeneral case with a set G of media di↵ering in their composition of amino acids.

Preparations A basal medium is prepared without amino acids, and distributedinto G flasks. Amino acids are added from stock solutions according to the experi-mental plan. The cell line is cultured in the reference medium, and the number ofcells is scaled-up in preparation for the parallel experiment.

Pseudo-perfusion protocol Cells from a common pool are seeded into TubeSpins,where the last TubeSpin corresponds to the control. A parallel pseudo-perfusion

75

Page 94: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

4. Experimental data: Influence of varied amino acid availability in CHO cellcultures

1h$1" 1h$2" 1h$3" 1h$G"...

...

$$$

1h$1" 1h$2" 1h$3" 1h$G"

Media

Parallel cultures

Data set

Data collection [t0 c0], [t c]

Input to programming software

Analytical work ...

...

Calculate rates [µ qmAb qext]

1 23

G

Figure 4.1: Workflow of a parallel cell culture experiment with media 1, 2, 3, . . . , G.

protocol is carried out with daily medium renewals (Figure 4.2). Parallel culturesavoid e↵ects due to cell history (Henry et al., 2008). In pseudo-perfusion, eachmedium renewal restores the nutrient levels and removes the by-products, providinga relatively stable environment for the cells. It is assumed that in each culture,a metabolic pseudo-steady state is reached within a matter of days, and that theexponential growth phase is maintained throughout the experiment. Each conditionis tested in a single culture, but each of those cultures are carried out for severalconsecutive days in the pseudo-perfusion mode1.

Sampling and data collection Samples are collected starting on day 4, and ana-lyzed with BioProfile FLEX, amino acid high-performance liquid chromatography

1See the study by De Jesus et al. (2004) for a comment about the reproducibility between duplicatecultures.

76

Page 95: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

4.2. Experimental plan

(HPLC), and IgG HPLC. The viable cell density, the cell viability and the concen-trations of glucose, lactate, glutamine, glutamate and ammonium are analyzed usinga BioProfile FLEX analyzer. The IgG concentration is analyzed with HPLC andthe concentrations of amino acids with amino acid HPLC. The resulting concentra-tion values are collected in spreadsheets, and read into the programming software.Cultivation data from one of the TubeSpin cultures are shown in Figure 4.2. Theviable cell concentration Xv is maintained at 2 · 106 viable cells/mL at each mediumrenewal and the viability is stable, slightly below 100 %, throughout the cultivation.Glucose and glutamine are taken up between each medium renewal, and lactateand ammonium are secreted. Data for two other amino acids are shown: glycine,omitted from the medium, is produced between each medium renewal and prolineis consumed.

Calculations The cell-specific rates of growth (µ) and uptake/secretion of ex-tracellular metabolites (qext) and mAb (qmAb) are calculated from concentrationmeasurements in samples collected before and after each medium renewal, accord-ing to the equations (2.1), (2.3), (2.4) in Chapter 2. The data set is now available formodel development.

4.2 Experimental planSixteen parallel cultures were planned for, of which the sixteenth was the controlof cells cultured in the original medium formulation. The concentration of a singleamino acid was varied in each of the remaining fifteen media, so that the e↵ectsobserved in each medium would be linked to a specific amino acid and concentrationlevel. The media are listed in Table 4.2; #1-15 are referred to by code Xy, where X isthe one-letter abbreviation of the varied amino acid and y is its concentration level inpercent relative to the concentration in the control medium #16 which was referredto as Ctrl. The availability of each amino acid was either a complete omission (0 %),a doubling (200 %), or a reduction (50 %).

Amino acids Among 20 potential amino acids (Table 2.9, p. 31), the ten aminoacids listed in Table 4.1 were selected for the experiment. Seven of those ten aminoacids are non-essential amino acids that can be synthesized by the CHO-K1 cells:alanine, asparagine, aspartate, glutamate, glutamine, glycine and serine (Table 2.5,p. 26). One is an amino acid for which the cells are auxotrophic (proline), one is

77

Page 96: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

4. Experimental data: Influence of varied amino acid availability in CHO cellcultures

Cell"pellet"

Cell*density*at*t"Discarded*volume"

Supernatant"

Sample*at*t"

Discard*Sample* Centrifuge* Sample* Re9suspend* Sample*

Cells*grow*over*night,*take*up*nutrients*and*secrete*by9products*

Sample*at*t0"

2*MVC/mL*

New*medium"

1- 2- 3- 4- 5- 6-

7-

t (day)0 2 4 6 8 10 12X

v (1

06 v

iab

le c

ells

/mL

)

0

1

2

3

4

5

Via

bili

ty (

%)

0

20

40

60

80

100

ViabilityX

v

t (day)0 2 4 6 8 10 12

mM

0

20

40

60GlcLac

t (day)0 2 4 6 8 10 12

mM

0

5

10

15

GlnNH

4+

t (day)0 2 4 6 8 10 12

mM

0

0.5

1

1.5

2

GlyPro

Figure 4.2: Daily TubeSpin pseudo-perfusion protocol (top) and example of data for thecase of glycine omission (bottom). The samples are collected just before and shortly afterthe re-seeding of the cells in new medium. The latter sample represents the start of eachrepetition at t0, and the former which is collected the next day represents the end at t. Here,Xv is the same as the viable cell density Cv, and Cv = 2 MVC/mL = 2 · 106 viable cells/mL.

definitely essential (threonine) and one is here assumed to be conditionally essential(cysteine can be obtained from methionine, see Table 2.7 on p. 29).

Omission of seven non-essential amino acids A complete omission was plannedexclusively for the non-essential amino acids alanine (A0), asparagine (N0), aspar-tate (D0), glutamine (Q0), glutamate (E0), glycine (G0), and serine (S0). As all20 amino acids are required for the synthesis of biomass and recombinant proteinproduct, intracellular synthesis of omitted non-essential amino acids was expected.A complete omission was favored over a reduction to: (i) promote the activation of a

78

Page 97: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

4.2. Experimental plan

Table 4.1: Ten amino acids to vary in the experiment.

Amino acid Three-letter One-letter Essentialityabbreviation abbreviation

Alaninea, e Ala A Non-essentialAsparagineb, e Asn N Non-essential

Aspartatee Asp D Non-essentialCysteinef Cys C Conditionally essential

Glutamatee Glu E Non-essentialGlutaminec,e Gln Q Non-essential

Glycined,e Gly G Non-essentialProlined Pro P AuxotrophySerinee Ser S Non-essential

Threonined Thr T Essentiala Addition of extra alanine to culture media can potentially inhibit the reaction of glutamate andpyruvate to alanine, and prevent secretion of alanine (Chen and Harcum, 2005).b Asparagine starvation (an immediate removal of asparagine from the culture medium) wasfound to influence extracellular and/or intracellular pools of several amino acids and increase thegrowth rate of CHO-DUKX cells (Seewöster and Lehmann, 1995). As seen in Table 2.9, thecells first took up asparagine from the medium. After the asparagine removal, this uptake shiftedtoward secretion.c Glutamine is a non-essential amino acid typically consumed by CHO cells at high rate con-tributing to extensive by-product secretion (p. 25). The impact of varied glutamine availabilityon CHO cell metabolism has been demonstrated (Wahrheit et al., 2014a), and is indicated inTable 2.9.d Addition of extra proline, threonine or glycine to the culture medium was found to improve thegrowth and the recombinant protein productivity and quality in CHO cell cultures subjected toelevated ammonium levels simulated by addition of NH4Cl (Chen and Harcum, 2005).e Non-essential and synthesis should be possible.f Conditionally essential and synthesis could be possible from methionine (assumed here).

synthetic pathway; and (ii) avoid depletion of the amino acid between the time pointsof sample collection. In the case of glutamine omission (Q0), a reduction in thegrowth rate was expected. Glutamine synthesis should be possible via asparaginesynthetase and glutamine synthetase (reactions 35 and 36 in Table 2.5, p. 26).

Zero initial concentrations (0 mM) were thus present by design, which is rare.In medium development, this represents a scenario in which a medium componenthas been purposefully omitted. During the course of a cell culture, very low or zeroconcentration levels can also be encountered due to depletion of substrates.

79

Page 98: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

4. Experimental data: Influence of varied amino acid availability in CHO cellcultures

Table 4.2: The selected concentration levels in the experimental media.Ctrl is the control.

# Amino acid Level Code # Amino acid Level Code(%) (%)

1 Alanine 0 A0 9 Glutamate 0 E02 Alanine 200 A200 10 Glycine 0 G03 Asparagine 0 N0 11 Threonine 200 T2004 Asparagine 200 N200 12 Serine 0 S05 Aspartate 0 D0 13 Serine 200 S2006 Aspartate 200 D200 14 Proline 200 P2007 Glutamine 0 Q0 15 Cysteine 50 C508 Glutamine 200 Q200 16 — — Ctrl

Doubling of five non-essential amino acids Five non-essential amino acids weredoubled in concentration: alanine (A200), aspartate (D200), asparagine (N200),glutamine (Q200), and serine (S200). A high initial concentration of e.g. alanine,aspartate or glutamate could inhibit their secretion as ammonium sink by-productsof the glutaminolysis (p. 25). A high initial concentration of either asparagine orglutamine could contribute to increased uptake of the amino acid, and additionalammonium formation.

Doubling or reduction of (conditionally) essential amino acids In the case ofthreonine, an essential amino acid, the concentration was doubled (T200). TheCHO-K1 cell line cannot synthesize proline (Wurm and Hacker, 2011), and therebythe concentration of proline was doubled (P200) rather than omitted or reduced. Inthe case of cysteine (C50) the concentration was halved rather than doubled to avoidtoxic e↵ects (Nishiuch et al., 1976) or omitted since cysteine is practically essentialin CHO cell culture applications (Sigma-Aldrich Media Expert).

4.3 Experimental dataThe data generated in the experiment consisted of the cell-specific rates for thegrowth, IgG secretion and uptake/secretion of 21 metabolites. Plots of the dataare found in Figures 2, and 4-6 of Paper I. In brief, the CHO-K1SV cell linegrew and produced IgG in all the sixteen media. Aside from the amino acids thatwere deliberately omitted at the initiation of the experiment, none of the measuredmetabolites were depleted. Variations in the growth, IgG production, and metabolite

80

Page 99: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

4.3. Experimental data

uptake/secretion rates were registered in response to changes in the amino acidavailability.

4.3.1 Control

The control culture serves as a reference within the experiment. Here, the cellsdisplayed a behavior typical for CHO cell lines in glucose- and glutamine sup-plemented medium: a growth rate of ⇡ 0.6/day; high-rate uptake of glucose andglutamine; high-rate secretion of lactate and ammonium; uptake of essential aminoacids isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan,and valine; and uptake of proline for which the CHO-K1SV cell line is auxotroph.Conditionally essential amino acids tyrosine and cysteine were taken up. Non-essential alanine, aspartate, glutamate, and glycine were secreted, while asparagineand serine were taken up.

4.3.2 Influence of varied amino acid availability

The varied availability of asparagine, aspartate, glutamine, glycine, serine andcysteine were associated with significant di↵erences in the metabolic rates, whilevaried availability of alanine, glutamate, proline and threonine had a smaller impact.Some observations will be described below.

Concerning the growth, viability, IgG production, glucose uptake and lactatesecretion, the cells showed similar behavior across all test media, with one exception:cells subjected to glutamine omission in Q0. The omission of glutamine from theoriginal medium led to a dramatic decrease in the cell-specific growth rate and theviability. In contrast, the cell-specific IgG secretion rate was dramatically increased.No glutamine was detected in Q0; the glutamine produced appears to have beenkept intracellularly. The cell-specific glucose uptake and lactate secretion rateswere dramatically increased compared to the control, while the secretion rate forammonium was decreased. Thereby glutamine uptake appears, at least in part, havebeen compensated for by an increased uptake of glucose in this medium.

Next, for the essential amino acids isoleucine, leucine, lysine, methionine,phenylalanine, threonine, tryptophan, and valine, and conditionally essential tyro-sine, the cell-specific uptake rates were similar across all the media with exceptionfor Q0. In Q0, these rates were generally increased compared to the control and theother media, in particular the uptake rate for isoleucine, leucine and methionine.

Asparagine and glutamine were consumed by the cells in the control medium,and in all the other media except N0 and Q0. In N0, no asparagine could be detected.

81

Page 100: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

4. Experimental data: Influence of varied amino acid availability in CHO cellcultures

Similarly, in Q0 no glutamine could be detected. Thereby, in these two cases, theamino acid appears to have been produced (as it is required in biomass- and productsynthesis) and kept intracellularly.

Alanine, aspartate, glycine and glutamate were produced by the cells in thecontrol medium. The omission of alanine, aspartate, glycine and glutamate wasexamined in the experiment, as was the doubling of alanine and aspartate. In eachcase, the omitted amino acid was produced and secreted, just as in the control, butin some cases at an altered cell-specific rate. For the cells subjected to variation inthe availability of aspartate, the secretion rate was increased when the amino acidwas omitted (D0) and decreased when the amino acid was doubled in concentration(D200). For the cells in G0 (glycine omission), the secretion rate was increasedcompared to the control.

In the control medium, a small amount of serine was consumed, consistent withthe typical pattern observed in mammalian cell cultures (Gódia and Cairó, 2006).In S0, serine was produced; thereby, its omission appears to trigger its productionand secretion. In G0, the uptake of serine occurred at an increased rate comparedto the control culture. In the control, cysteine was consumed. In C50, the specificconsumption rate appeared slightly lower than in the control, and in most of theother media.

The next chapter takes as the starting point the generation of a macroscopickinetic model to capture these data, with the objective to simulate the complete dataset using a single model.

82

Page 101: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Chapter 5

Contribution of Paper I:Development of an initialmethodology

The aim in Paper I was the same as in the thesis. This chapter begins with anintroduction to the proposed modeling concept, followed by a description of theoverall workflow (Section 5.1). The following section focuses on the application anddevelopment of methodology, leading to a proof-of-concept (Section 5.2). Finally,Section 5.3 summarizes the main contributions of the paper.

5.1 Proposed concept and workflowThe task in Paper I was to find a way to generate a single model that could simulate adata set encompassing sixteen di↵erent experimental conditions over several culturedays. At the time point of the study, measurements of 22 di↵erent variables wereavailable, i.e., the cell density, and the concentrations of glucose, lactate, ammonium,and eighteen natural amino acids.

5.1.1 Modeling concept

A one model approach was envisioned in which a single model M would be devel-oped from the entire data set (Figure 5.2). With the methodology in Section 3.7 as thestarting point, it was proposed that all available data were to be used simultaneously

83

Page 102: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5. Contribution of Paper I: Development of an initial methodology

within that framework to yield a single metabolic model. To capture diverse behaviorof the cell line, that model was to rely on the following principles:

• Principle 1: The cells have access to several pathways that link the extra-cellular metabolites; in the model those are the EFMs of the metabolic reactionnetwork.

• Principle 2: An observed metabolic behavior of net uptake/secretion ratesresults from a combination of pathway activities; in the model this relation-ship is captured by stating an EFMs-based MFA problem that describes adecomposition of observed behavior qext into pathway activities w (see p. 62).

• Principle 3: The cells regulate those pathway activities in response to environ-mental changes, and the regulation is thought of as the action of switchingon/o↵, or increasing/decreasing the pathway flux; in the model, the regulationof each pathway activity wl is modeled via kinetic functions of the typewl = wmax,l · fl(c1, c2, ..., k1, k2, ...).

A conceptual illustration is shown in Figure 5.1.

I

G

C

B

FD

A

H

J

E

w

w

w

w

I

G

C

B

FD

A

H

J

E

w

w

w

w

I

G

C

B

FD

A

H

J

E

Figure 5.1: A conceptual illustration of the three principles for the metabolic model. Aset of possible pathways through the cell metabolism indicated by dashed lines (left). Twodi↵erent pathway flux distributions (middle and right) for which the switching on/o↵ of apathway is indicated by colored lines (turned on) versus dashed lines (switched o↵), and theincrease/decrease of pathway flux by variation in the line thickness.

5.1.2 Workflow toward proof-of-concept

The workflow is outlined in Figure 5.3. The workflow strived toward proof-of-concept, meaning a realization of the modeling concept that demonstrates its feasi-

84

Page 103: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5.1. Proposed concept and workflow5.1. Proposed concept and workflow

A

B

C

D

A

B

C

D

?

Multiple models One modelScenarios Models Simulations Scenarios

Model

Simulation

?E

New scenario Prediction

?

New scenario Prediction

? ? ? ?

Unknown optimal scenario

Unknown optimal response

Unknown optimal scenario

Unknown optimal response

M

A

B

C

D

A

B

C

D

A

B

C

D

M

E

Figure 5.2: Conceptual illustration of a one-model approach. A model M is developedfrom input-output data. Prediction: for a new scenario X with input c0,X , the model predictsthe output qext,X . Optimization: the model finds an optimal medium composition c⇤0 thatyields the best output behavior in terms of specified objectives and constraints. Note thatprediction and optimization were outside the scope of the paper but are long-term goals.

bility and potential, but may also point out limitations and challenges that should besolved in future work.

While the workflow resembles the one in Section 3.7, that methodology is nowapplied to a specific situation. The experiment (1) is the parallel culture experimentin Chapter 4. As before, the user specifies a metabolic reaction network (2). This isfollowed by the usual enumeration of EFMs by Metatool (3), and a translation ofthose EFMs into the macroscopic reactions (4). Next, the entire data set of sixteenconditions is inserted into the EFMs-based MFA problem (5). In practice, this isachieved via stacked matrices and vectors1. The user then specifies a set of kineticfunctions and parameters (6); those are initially based on general equations withsubstrate saturation e↵ects. The parameter estimation (7) is limited to wmax, whereeach metabolite has a fixed saturation parameter value applied across all equations.Finally, and as before, the model is evaluated based on the quality of the data fitting(8).

As a new feature, there are now three feedback loops A, B and C in the work-1 See Section 4.3.1, Paper I

85

Simulate

Predict

Optimize

5.1. Proposed concept and workflow

A

B

C

D

A

B

C

D

?

Multiple models One modelScenarios Models Simulations Scenarios

Model

Simulation

?E

New scenario Prediction

?

New scenario Prediction

? ? ? ?

Unknown optimal scenario

Unknown optimal response

Unknown optimal scenario

Unknown optimal response

M

A

B

C

D

A

B

C

D

A

B

C

D

M

E

Figure 5.2: Conceptual illustration of a one-model approach. A model M is developedfrom input-output data. Prediction: for a new scenario X with input c0,X , the model predictsthe output qext,X . Optimization: the model finds an optimal medium composition c⇤0 thatyields the best output behavior in terms of specified objectives and constraints. Note thatprediction and optimization were outside the scope of the paper but are long-term goals.

bility and potential, but may also point out limitations and challenges that should besolved in future work.

While the workflow resembles the one in Section 3.7, that methodology is nowapplied to a specific situation. The experiment (1) is the parallel culture experimentin Chapter 4. As before, the user specifies a metabolic reaction network (2). This isfollowed by the usual enumeration of EFMs by Metatool (3), and a translation ofthose EFMs into the macroscopic reactions (4). Next, the entire data set of sixteenconditions is inserted into the EFMs-based MFA problem (5). In practice, this isachieved via stacked matrices and vectors1. The user then specifies a set of kineticfunctions and parameters (6); those are initially based on general equations withsubstrate saturation e↵ects. The parameter estimation (7) is limited to wmax, whereeach metabolite has a fixed saturation parameter value applied across all equations.Finally, and as before, the model is evaluated based on the quality of the data fitting(8).

As a new feature, there are now three feedback loops A, B and C in the work-1 See Section 4.3.1, Paper I

85

Figure 5.2: Conceptual illustration of a one-model approach. A single model M is devel-oped from input-output data of multiple medium scenarios A, B, C, and D. Prediction: fora new scenario E with input c0,E , the model predicts the output qext,E . Optimization: themodel finds an optimal medium composition c⇤0 that yields the best output behavior in termsof specified objectives and constraints. Note that prediction and optimization were outsidethe scope of the paper, but are long-term goals.

bility and potential, but may also point out limitations and challenges that should besolved in future work.

While the workflow resembles the one in Section 3.7, that methodology is nowapplied to a specific situation. The experiment (1) is the parallel culture experimentin Chapter 4. As before, the user specifies a metabolic reaction network (2). This isfollowed by the usual enumeration of EFMs by Metatool (3), and a translation ofthose EFMs into the macroscopic reactions (4). Next, the entire data set of sixteenconditions is inserted into the EFMs-based MFA problem (5). In practice, this isachieved via stacked matrices and vectors1. The user then specifies a set of kineticfunctions and parameters (6); those are initially based on general equations withsubstrate saturation e↵ects. The parameter estimation (7) is limited to wmax, whereeach metabolite has a fixed saturation parameter value applied across all equations.Finally, and as before, the model is evaluated based on the quality of the data fitting(8).

1 See Section 4.3.1, Paper I

85

Page 104: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5. Contribution of Paper I: Development of an initial methodology

As a new feature, there are now three feedback loops A, B and C in the work-flow. These have been introduced here to indicate iteration procedures, and theirrelevance will become evident in Section 5.2. First, Feedback A connects the EFMenumeration (3) to the network structure (2). Adjustments at 2 can be done toensure that the EFMs can be enumerated by Metatool at 3. Furthermore, if theenumeration is feasible, the applied network structure also determines the set ofEFMs and macroscopic reactions that will be available for the kinetic model de-velopment. Next, Feedback B connects the evaluation (8) to the network structure(2), and finally feedback C connects the same evaluation (8) to the introduction ofkinetic functions and parameters (6). Within this workflow, it is thereby possible toexplore if certain adjustments of the network and/or the kinetic functions/parametersimprove or worsen the quality of the data-fitting; repeated iterations guide the usertoward a proof-of-concept model.

5.2 Application and further developmentThe overall objective of the work was to reach the proof-of-concept model at thebottom of the workflow, and the actual model is presented in more detail in the paper.But the work also led to new insights about the concept, and to further developmentof the methodology itself; these will be discussed in the present section. The sectionfocuses on two important aspects: (i) the specification of a suitable network andidentification of its EFMs (Section 5.2.1); and (ii), finding suitable kinetic functionsand estimating their parameters (Section 5.2.2). Lastly, the proof-of-concept modelpresented in Paper I will be briefly discussed.

5.2.1 Specification of a suitable network and identification of itsEFMs

In terms of reactions, the amino acid metabolism should logically be representedin the network: essential and non-essential amino acid catabolism; synthesis ofnon-essential amino acids; and, use of amino acids in the biomass and productsyntheses. Additionally, the diverse behavior displayed in the data called for aflexible network structure. This implied that shifts in the net direction of severalreactions should be allowed (see p. 26), including reversible transports.

The first networks to enter the workflow (2) were of similar size and detail asother cell culture models available in the literature at the time (p. 68); but, given thetopic of the study, reactions that provided a more complete coverage of the aminoacid metabolism were included, and many of the network reactions were defined

86

Page 105: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5.2. Application and further development

1Parallelculture

experiment

2Specify metabolicreaction network

3Enumerate EFMs

4Translate intomacroscopic

reactions

5State the EFMs-

based MFA problem

6Introduce

kinetic functions

7Estimate the

parameters andsimulate rates

8Evaluate

Proof-of-concept model

qext

qext

c0

K

Aint, virrev

E, eirrev

Amac

w

fl

wmax, qsim

Aext

A

C

B

Figure 5.3: Workflow toward proof-of-concept in Paper I, where qsim refers to the simulatedrates. The execution of steps 3–5, and 7 was mostly computational, whereas steps 1, 2, 6,and 8 at this point required human involvement and decision-making.

87

Page 106: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5. Contribution of Paper I: Development of an initial methodology

as reversible. Unfortunately, the EFMs of the preliminary networks could not beenumerated, as the Metatool algorithm terminated before the enumerations werecompleted.

For pragmatic reasons, less complex networks had to be tested within theframework to arrive at proof-of-concept. The procedure involved tradeo↵s be-tween various network properties, mostly related to internal nodes and reactionreversibility.

Revisiting the workflow outlined in Figure 5.3, network adjustments that makethe EFM enumeration feasible are possible via feedback A, and the influence ofthose adjustments on the quality of the data fitting can be accounted for via feedbackB.

In the end, as can be seen in the map of the proof-of-concept network (Figure 5.4),the NEAA metabolism and its internal nodes and interconversions were prioritized,as was the reversibility of related reactions and transports. When provided with thisnetwork, the Metatool algorithm enumerated 379 EFMs. Unfortunately, this cameat the cost of loosing several of the relevant internal nodes, and unfortunately alsothe EAA catabolism.

5.2.2 The kinetics

For the kinetic model development, the following equation served as the startingpoint,

wk,l = wmax,l ·Y

i2Mext,s,l

c0,k,iKs,i,l

c0,k,iKs,i,l+ 1

| {z }substrate saturation

,(5.1)

a version of (3.18) where k has been introduced to account for the stacking of data2.In its favor, a generalized equation of this type is convenient for automatic modelgeneration because every function can be derived directly from Amac. Nevertheless,a saturation parameter for each extracellular substrate still has to be specified andfixed beforehand.

As previously described, the balanced growth assumption has been appliedin the literature. When substrates are available in excess, this entails that mostsaturation parameters can be fixed to low values (p. 68). An example plot isshown in Figure 5.5. Clearly, the condition of excess substrate availability was notapplicable in the present case; metabolites had in fact been omitted by design. In

2 See Section 4.3.1, Paper I

88

Page 107: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5.2. Application and further development

Lacext

Glcext

AcCoA

Pyr

Mal

Oxal

SucCoA Suc

Pheext

Tyrext

Ileext Metext Thrext Valext

Gln Glu

Leuext Pheext Trpext Lysext Ileext Thrext Tyrext Pyr Ala

Proext Asp

Asn

aKG

Glu

Gly

Ser

Cysext

aKG

Gln

Glu

aKG Glu

Gluext

NH4+

ext

NH4+

ext

NH4+

ext

NH4+

ext

NH4+

ext

Glnext

Asnext

Aspext

Glyext

Serext

Alaext

3PG

Biomassext Pheext Tyrext Leuext Trpext Lysext Ileext Thrext Metext Valext

1"

2"

3"

4"

5"

6"

7"

8"

9"

10"

11"

12" 13"

14"

16"

17"

19"20"

21"

22"

24"

25"

26"

15"

27"

29"

30"

28"

23"NH4

+ext

18"

Cysext Proext

Alaext Glyext Serext Gluext Glnext Aspext Asnext

Figure 5.4: Metabolic network in Paper I. Eighteen reactions are reversible, includingseven of the transport reactions. NEAA metabolism is included, but the EAA catabolismwas excluded from the network and is here only indicated (green). This same map is foundin Figure 3 of Paper I, where the network is described in Section 4.2.1.

addition, note that in (5.1) and in Figure 5.5, if one substrate is omitted or depletes,the flux over the pathway is switched o↵, perfectly in line with fundamental enzymekinetics. This will however have consequences on the modeling of the globalbehavior.

A noteworthy aspect was the strong influence of the network structure on thekinetics. As a consequence of the generalization in (5.1), the EFMs of the networkfully determined whether the concentration of a certain metabolite could influence amodel at all (it had to be a substrate of a pathway), and also its potential interactionwith other metabolites (those had to be substrates in the same pathway).

An important insight was that even if a metabolite had been omitted or reducedin concentration at the initiation of a cell culture, from a global perspective, com-pensating pathways that produced it, and pathways that consumed it, could havebeen activated once that cell culture had reached a stable pseudo-steady state. As

89

Page 108: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5. Contribution of Paper I: Development of an initial methodology

an example, while the omission of serine triggered its own production, an initialserine concentration of zero could not activate or increase the flux over serine-producing pathways, only switch o↵ fluxes over pathways in which the amino acidwas consumed. This led to a first desired property:

• Desired property 1: Omission or reduction of an amino acid should be ableto activate and up-regulate a pathway activity.

Feedback A (in Figure 5.3) provides the option to change the reversibility ofreactions in the network; this way, new metabolites could enter the set of substrates.Nevertheless, the following situation always remained: (i) extracellular productscould not regulate the activity of a pathway; and (ii) an extracellular metabolitecould not regulate any of the pathways in which that metabolite was not involved.This observation led to a second desired property:

• Desired property 2: Non-substrate metabolites should be able to regulate apathway flux.

Furthermore, a model within the framework should not rely completely on theuse of small parameter values given the third principle of the metabolic model(p. 84). It entails that the cell line is able to increase or decrease a pathway fluxin response to environmental changes, as is illustrated by the plotted function tothe right in Figure 5.5, and via the two conceptual flux maps with di↵erent linethickness in Figure 5.1.

As a result, the following generalized equation was proposed:

wk,l = wmax,l · fl(c0,l,k,Kl),

where

fl(c0,l,k,Kl) =Y

i2Mext,s,l

c0,k,iKs,i,l

c0,k,iKs,i,l+ 1

| {z }substrate saturation

·Y

i2Mext,p,l

1c0,k,iKp,i,l+ 1

| {z }product inhibition

·Y

i2Mext,r,l

1c0,k,iKr,i,l+ 1

| {z }metabolite inhibition

.(5.2)

As indicated, the new equation consists of three terms to represent substrate satura-tion, product inhibition, and general metabolite inhibition. The equation has bothdesired property 1 and 2. The omission or reduction of a product can trigger its ownproduction through an inhibitory e↵ect, and presence of a general metabolite caninhibit the activity of pathways in which it is not involved.

90

Page 109: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5.2. Application and further development

S ext

w

Ks = 0.001

"Switch o↵"

w ⇡ wmax

0 1 2 30

1

2

3

4

S ext

w

Ks = 1

w varies

0 1 2 30

1

2

3

4

Figure 5.5: A plot of the equation w = wmaxS ext

(S ext+Ks)where wmax = 4 and Ks = 0.001 (left)

and Ks = 1 (right). S ext is the initial concentration of a single extracellular substrate, and wis the macroscopic flux rate. The shaded area between the dashed lines indicates a range ofinitial concentrations between 0.5 and 3.5 mM.

While it is true that (5.2) has the desired properties, it also yields a high numberof possible combinations. For the kinetic function of each macroscopic reactionmust be determined: the presence of saturation and/or inhibition, the metabolitesinvolved in those regulations, and the kinetic parameter for each case. FeedbackC allows a user to manually adjust the kinetics of each function to improve thedata-fitting, but the procedure quickly becomes tedious.

To speed up the identification of suitable kinetics, the following strategy wasdeveloped. A first model based on (5.1) served as a starting point. Based on itsability to fit the data, the user in this case adds new equations on the form (5.2).The result is an equation pool; each macroscopic reaction may be associated withmultiple equations. The subsequent parameter estimation assigns higher weightto certain equations in the pool (Figure 5.6). The procedure is repeated until asatisfactory model has been obtained.

The new general equation (5.2) in combination with the above strategy wasreferred to as "flexible kinetics", where the decision on the equations to add isguided iteratively by the data-fitting. A low-quality fit of a certain flux gives a clueto what type of equation and parameter values that could be useful. The structuralinformation available in the EFMs and macroscopic reactions is applied to findsuitable connections between metabolite fluxes and metabolite concentrations (seeTable 3 in Paper I), and the direction of the mis-match can guide the adjustment of

91

Page 110: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5. Contribution of Paper I: Development of an initial methodology

the model parameters.

Equation poolInitialmodel

Reducedmodel

Estimate Sort Reduce

CF

1A1B2A2B2C

34

5A5B

67A7B7C

3

7D8A8B

910

Pathways

123456789

10

Kinetics

2B6

1A5B7B2A

45A7A

32B

61A5B7B2A

4

7B8A

1 2 3 4

109

1B8B7D7C

Figure 5.6: Conceptual illustration of a data-driven selection of the kinetic functions. Thered boxes illustrate a simple case with the ten pathways #1–10 identified via an EFMenumeration (and which have been translated into their corresponding ten macroscopicreactions). At step 1, a pool of kinetic equations is formulated, where it is possible to defineseveral alternative equations for each pathway; here this is indicated by letters A, B, C etc.At step 2, the linear parameter estimation yields a parameter wmax,l for each equation, andsome equations will be associated with a higher parameter value/weight in the model; herethe weight is indicated by an increasingly darker blue color.

5.2.3 Proof-of-concept model

A proof-of-concept model was obtained through numerous iterations within theworkflow. The simulations are available in Figures 4-6, Paper I.

From those simulations can be seen that the model has an ability to capturetypical CHO cell behavior, as the rates of the control culture are overall wellsimulated. The underestimation of essential amino acid uptake is an exception(Figure 6, Paper I), but must be expected, given the exclusion of EAA catabolism tofavor flexibility in the NEAA metabolism.

In line with the aim, the ability of the same model to simulate diverse cell linebehavior is also demonstrated; many of the variations in the uptake/secretion ratespresent in the data set are well captured (see specifically Figures 4 and 5, Paper I).This indicates that the modeling concept has potential for the generation of singlemodels with ability to capture diverse cell line behavior.

92

Page 111: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

5.3. Summary of the contribution

5.3 Summary of the contributionTo conclude, the main contribution of Paper I is a proposal and demonstrationof a new concept for how to generate a single cell culture model, with ability tosimulate diverse behavior of a specific cell line triggered in response to changesin medium composition. Even if macroscopic kinetic models based on metabolicreaction networks and simplified kinetics have been suggested previously in theliterature, the paper demonstrates an application of the model type to a new situation,i.e. a situation in which a cell line is cultured in parallel pseudo-perfusion culturesrather than in a single batch culture, and subjected to sixteen di↵erent mediumcompositions rather than to a single medium.

The amino acid metabolism in CHO cells is characterized by metabolic flexibil-ity (p. 28), and the experiment in Chapter 4 was purposefully designed to triggeractivation of alternative and compensatory pathways within this metabolism. Clearly,the modeling of diverse behavior requires a more flexible modeling structure thancan be provided by simplified networks and simple generalized kinetics.

As proof-of-concept, the paper demonstrates feasibility and potential of thesuggested modeling concept via the realization of an actual model. But it alsocontributes to new insights and points out challenges that should be sorted outin future work. An important finding was the limitation imposed by the EFMenumeration step (3); only simplified networks could be used, which requiredsacrifices of desired network properties. This limitation was formulated as a newproblem to be solved, and was handled in Paper II which will be described in thenext chapter. Another finding was the limitation of generalized substrate saturationkinetics, something which could be solved in part through a new flexible equationin combination with a strategy for selecting kinetics for the model. Additional workon the kinetics will be presented in Chapter 7 (Paper III).

93

Page 112: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work
Page 113: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Chapter 6

Contribution of Paper II: Acolumn generation-based methodfor pathway identification incomplex metabolic networks

In Paper II, the issue of pathway identification in complex metabolic networkswas considered. To address the issue, a novel method based on a mathematicaloptimization technique referred to as column generation (CG) was proposed. Thischapter begins with an introduction to mathematical optimization, linear programm-ing, and the CG technique (Section 6.1). Section 6.2 describes the proposed methodand its implementation, and Section 6.3 summarizes the main contributions of thepaper.

6.1 Introduction to mathematical optimizationOptimization problems are found in various fields, e.g. in manufacturing, transporta-tion, finance, and mechanics, as well as in the biological sciences. The commonfeature of these problems is the aim to select a best solution to the problem froma set of candidate solutions. An objective function is the output to be maximizedor minimized; it quantifies the quality of each solution. Examples could be tomaximize profits or to minimize waste, or as in the present case to optimize thequality of a data-fitting through the minimization of the sum of the squared residuals

95

Page 114: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

6. Contribution of Paper II: A column generation-based method for pathwayidentification in complex metabolic networks

between a model and data. Constraints are requirements that must be fulfilled, andtypically expressed as equalities or inequalities.

6.1.1 Linear programming (LP)

Linear programming (LP) are techniques for solving optimization problems basedon linear relationships: a linear objective function, and linear constraints. Thestandard form of such linear programs (LPs) is to,

minimizex

cT x,

subject to Ax = b,x � 0,

(6.1)

where b and c are known vectors, and A is a known matrix (Wallis, 2012). LPalgorithms are designed to find an x that yields the smallest value of the objectivefunction cT x, if such a value exists. If it does not exist, the problem is infeasible orunbounded with no solution. Every LP can be re-written into a corresponding dualproblem, where the primal problem is the original LP. The variables, constraints andoptimal solutions of the primal and dual problems are related to each other as: eachdual variable corresponds to a primal constraint; each primal variable correspondsto a dual constraint; and an optimal solution to the dual problem can be recoveredfrom an optimal solution to the primal problem, and vice versa (Arora, 2017). Thisway, only one of the two problems needs to be solved, which can prove useful insome situations.

6.1.2 Column generation (CG)

The CG technique is a technique developed to e�ciently solve large LPs in which xconsists of a huge number of variables. Its development can be traced back to the1950s (Table 6.1). The technique is particularly useful when many of the variablesin x tend to be zero during the solution process, and in the final optimal solution(Ford and Fulkerson, 1958). It is not necessary to know all the variables beforehand,and some of those variables will never be dealt with; rather than to identify allvariables in x, variables are identified as needed (Dantzig and Wolfe, 1960; Gilmoreand Gomory, 1961, 1963). This saves a considerable amount of time and memoryin the implementation, and the problem can be solved e�ciently. This e�ciencymakes CG a very powerful technique with applications in e.g. crew schedulingfor air tra�c, or in vehicle routing for delivery of goods (Desrosiers et al., 1984;Lübbecke and Desrosiers, 2005).

96

Page 115: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

6.2. Column generation applied to EFM identification

Table 6.1: Important contributions to the development of the column generation (CG)technique.

Year Authors Contribution1958 Ford and

FulkersonIn the maximum multi-commodity network flow problem,all variables do not need to be explicitly stated, but can bedealt with implicitly.

1960 Dantzingand Wolfe

Application of the Dantzig-Wolfe decomposition toextend the LP column-wise, and as needed, in the solutionprocess.

1961,1963

Gilmoreand Gomory

Developed an e�cient heuristic algorithm to solve thecutting-stock problem. Showed that the problem couldbe e�ciently solved to optimality by using an MP andSP combination, without prior knowledge of all possiblesolutions.

1984 Desrosierset al.

Embedded CG techniques within a LP-based branch-and-bound (BnB) framework to solve a complex vehicle rout-ing problem.

In CG, an LP is split into a Master problem (MP) and one or several subproblems(SPs) (Dantzig and Wolfe, 1960; Gilmore and Gomory, 1961, 1963). The MP issimilar to the original LP, but it only contains the variables in x that are active (i.e.non-zero). The SP is referred to as the column generator (Lübbecke and Desrosiers,2005); during the solving process, it identifies new candidate variables for the MP,and specifically candidates that lower the MP objective function.

6.2 Column generation applied to EFM identificationRevisiting the EFMs-based MFA-problem (3.12) on p. 62, but in a version thataccounts for the stacking in Paper I,

minimizew

12

DX

k=1

kqk � IkAextEwk22

subject to w � 0,

97

Page 116: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

6. Contribution of Paper II: A column generation-based method for pathwayidentification in complex metabolic networks

it is assumed that the full matrix E is known from an a priori EFM enumerationbefore the problem can be solved. Unfortunately, when the complexity of the under-lying network increases, the number of EFMs increases rapidly, and the enumerationquickly becomes prohibitive. This is a general issue in EFM enumeration (Klamtand Stelling, 2002) and was also identified as a limitation in the kinetic frameworkproposed in Paper I.

6.2.1 Proposition

Column generation emerged as suitable mathematical technique to solve the issuebecause: (i) (3.12) is a large LP with a high number of variables in w when thenetwork is complex; (ii) E can be recognized as a corresponding large set of columns,unknown beforehand; and (iii) if there exists one or several optimal solutions w inwhich many of the fluxes are zero, there is a chance that the problem could be solvede�ciently using the CG technique, and without an a priori EFM enumeration. Anapplication of the CG technique was therefore proposed to solve (3.12). The EFMswould not have to be enumerated, but identified as needed to arrive at an optimalsolution in terms of the quality of the data-fitting.

6. Contribution of Paper II: A column generation-based method for pathwayidentification in complex metabolic networks

E # # # # # # # # # # # . . . # # #

# # # # # # # # # # # . . . # #

EB # # # # # # # # # # # # # # # #

| {z }EB

e

Figure 6.1: The columns represent EFMs of a network. The upper row of columnsillustrates a large number of EFMs in E, enumerated for a complex network. Col-umn generation applied to the EFMs-based MFA problem aims to identify a subsetof those EFMs, EB (and a flux vector wB), that can fit a data set optimally. The rowof columns below illustrates the ongoing identification of the EFMs in that subsetEB. The red columns are the EFMs currently included in the subset EB. The orangecolumn is a candidate EFM from e that is under evaluation. If the inclusion of thiscandidate EFM improves the objective function, it will be added to the subset EB.

v = Ew = EBwB (6.2)

][The green columns illustrated in the Figure represent EFMs (these are vectors

of type el, which make up matrices E and EB). The blue boxes represent fluxesw (these are contained in vector w) As was explained in Section X, the time andcomputational power required for a complete enumeration of all possible EFMsbecomes prohibitive for large and complex networks.]

6.4.1 Master problem (MP) and subproblem (SP)

The EFMs-based MFA problem in which E has been replaced by EB is transformedinto a column generation problem by defining the master problem (MP) and a suit-

100

?

Figure 6.1: A split of the EFMs of a network into: the EFMs associated with activevariables currently in EB (red); a candidate EFM under evaluation (orange); and the EFMsassociated with inactive variables wN currently in subset EN (green), most of which areunknown.

98

Page 117: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

6.2. Column generation applied to EFM identification

6.2.2 Method

The next step was to formulate an MP and SP combination that could solve (3.12).Hildur Æsea Oddsdottir (HÆ) is the contributor to this formulation which is thor-oughly described in Paper II and in her thesis (Oddsdóttir, 2015). The method isintroduced also in the present thesis to support its application in papers III and IV.Furthermore, when using an algorithm it is valuable to understand how the inputsare processed into the outputs, e.g. to determine if the method can be applied to aspecific problem, and to interpret the results.

Division into subsets The CG technique handles subsets of the columns in anunknown matrix. In the present case, the unknown matrix is the set of all possibleEFMs in E, with corresponding variables w. This matrix is split into two matrices,such that E = [EB EN] where EB contains the subset of columns handled explicitlyby the CG algorithm, and EN the remaining columns (Figure 6.1). Similarly, theunknown solution vector w is split into two vectors, w = [wB wN]T , where wBcontains the fluxes over the EFMs in EB, and wN the fluxes over the EFMs in ENwhere wN = 0.

The Master problem (MP) and subproblem (SP) In the CG technique, an MPis derived from the original LP, so in the present case an MP is derived from (3.12).As an MP only considers the active variables during the solution process, E isreplaced by EB, and w by wB, such that the MP is to,

minimizewB

12

DX

k=1

kqk � IkAextEBwBk22 (6.2a)

subject to wB � 0, (6.2b)

where EB is a set of known EFMs, and w⇤B will denote an optimal solution to theproblem. Noteworthy, at this stage, w = [w⇤B wN] = [w⇤B 0] is not necessarily anoptimal solution to the full problem (3.12). The optimality conditions for the MPand (3.12) are discussed in Section 3.1 of Paper II. The SP objective function was

99

Page 118: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

6. Contribution of Paper II: A column generation-based method for pathwayidentification in complex metabolic networks

6. Contribution of Paper II: A column generation-based method for pathwayidentification in complex metabolic networks

Network Data Bounds

Initialcolumn

EB=e

MP

ImprovingcolumnEB=[EB e]

w⇤B, �⇤B

w⇤Boptimal? SP

Iterationsstop

CG

Optimalsolution

EB, w⇤B

No

Yes

Figure 6.2: CG algorithm. Flow chart showing the CG-based identification of an EFMsubset using a CG-based algorithm. �⇤B are dual variables obtained from the MP to accountfor bounds on unmeasured extracellular metabolite uptake/secretion rates and are introducedinto the problem in Section 6.2.4.

based on the last optimality condition of the MP such that the SP was to:

minimizee

DX

k=1

(ATextIT

k (IkAextEBw⇤B � qk))T e (6.3a)

subject to Ainte = 0, (6.3b)

1T e 1, (6.3c)e � 0. (6.3d)

where EB is the current EFM subset in the MP, and w⇤B is the current optimal solutionto the MP. An optimal solution to the SP is denoted by e⇤. The objective (6.3a)

100

Figure 6.2: The flow chart shows the identification of an EFM subset using the CG–based algorithm. �⇤B are dual variables obtained from the MP to account for bounds onunmeasured extracellular metabolite uptake/secretion rates and are introduced into theproblem in Section 6.2.4.

based on the last optimality condition of the MP such that the SP was to:

minimizee

DX

k=1

(ATextIT

k (IkAextEBw⇤B � qk))T e (6.3a)

subject to Ainte = 0, (6.3b)

1T e 1, (6.3c)e � 0. (6.3d)

where EB is the current EFM subset in the MP, and w⇤B is the current optimal solutionto the MP. An optimal solution to the SP is denoted by e⇤. The objective (6.3a)

100

Page 119: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

6.2. Column generation applied to EFM identification

was chosen because: (i) when e⇤ yields a negative SP objective value, the additionof e⇤ to EB improves the quality of the data-fitting in the MP; and (ii) when theSP objective value is zero, w⇤B is a global optimal solution both to the MP andto the full problem in (3.12). The SP thus allows the algorithm to find promisingcandidate columns, and to determine if they should be added to the MP and whento stop the iterations (Figure 6.2). The functions of the constraints are to fulfill thepseudo-steady state assumption (6.5b), bound the problem (6.5c), and ensure thatthe reactions of the network are used irreversibly (6.5d).

The workflow of the CG-based algorithm is shown in Figure 6.2. As indicated,the algorithm starts from a single EFM e that forms the first EFM subset EB. Solvingof the MP generates a corresponding optimal solution w⇤B. Given EB and w⇤B, theSP generates an EFM whose addition to EB improves the MP (the SP objectiveis negative). These procedures are iterated until the current EB solves the MP tooptimality. This happens when the objective function of the SP is zero, and thealgorithm stops. The current EB and w⇤B delivered as output solves both the MPand (3.12) to optimality, and the solution is a global optimum to (3.12). There isno better solution in terms of the objective value, but note that the solution is notnecessarily unique; there may be other solutions equally optimal.

6.2.3 Implementation and case-study

The method was implemented in the Matlab environment, and a case-study servedas a test of the proposed method. The CG-based algorithm developed within thework was able to solve an EFMs-based MFA problem in practice, also in a case inwhich the a priori EFM enumeration was prohibitive.

Data from one of the cell cultures (G0, Chapter 4) were used. For a simplenetwork (N1 in Table 3.4), an enumeration by Metatool yielded 257 EFMs in around2 s, while the algorithm based on the CG technique identified 14 of those EFMsthat fit the data set optimally in around 7 s1. For a more complex network (N2 inTable 3.4), enumeration by Metatool was prohibitive, but the CG-based algorithmwas able to identify 25 EFMs, again in around 7 s. An important finding was thatthe complex network gave an improved data-fitting when compared to the simpleone, indicating practical value of the approach.

Figure 6.3 contrasts the approach relying on a priori EFM enumeration againstan EFM identification through the CG technique. In the first case, the EFM enumer-

1As noted in the paper, all calculations were carried out on a single personal computer (64-bitLinux, a single Intel Xeon 3GHz processor core, 32GB of memory) and the implementation was notoptimized in terms of run time.

101

Page 120: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

6. Contribution of Paper II: A column generation-based method for pathwayidentification in complex metabolic networks

Metabolicnetwork

Metatool

EFMs-basedMFA

problem

w⇤

Data qext

Aint, virrev

E, eirrev

Amac

Aext Metabolicnetwork

CGData

EB, w⇤B

qext

Aint, Aext, virrev

Figure 6.3: A priori enumeration of all possible EFMs in E using e.g. Metatool to statethe EFMs-based MFA problem (left), versus a proposed scenario in which a relevant EFMsubset EB is identified using the CG technique (right). In the first case (left), the EFMenumeration and the data-fitting are carried out in separate steps while in the second case(right), the EFM subset identification and data-fitting are carried out simultaneously withina CG-based algorithm.

ation and the data-fitting are carried out in two separate steps. In the second case, theEFM identification and data-fitting are carried out simultaneously. A substitution ofthe EFM enumeration in the kinetic framework (Figure 5.3) by the new CG-basedmethod can now be imagined. The application of the method in kinetic modelingwill be covered in Chapter 7, but already here it can be noted that while the newmethod solves the EFMs-based MFA problem for a single pseudo-steady state, itdoes not account for the multiple pseudo-steady states or the kinetics studied inPaper I.

102

Page 121: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

6.2. Column generation applied to EFM identification

6.2.4 Robustness and bounds on unmeasured metaboliteuptake/secretion rates

Paper V was excluded from this thesis, but presented an extension of the prob-lem in Paper II into a robust format that could handle measurement errors2. Anadditional feature was an option to include bounds on the unmeasured metaboliteuptake/secretion rates. This option was utilized in papers III and IV. The new MP(here without robustness taken into account) was to,

minimizewB,zUB,zLB

12

DX

k=1

kqk � IkAextEBwBk22 + MTUBzUB +MT

LBzLB (6.4a)

subject to wB � 0 (6.4b)

zUB � AnmEBwB � �qUBnm (6.4c)

zLB + AnmEBwB � qLBnm (6.4d)

zUB � 0 (6.4e)

zLB � 0. (6.4f)

and the new SP was to,

minimizee

DX

k=1

((ATextIT

k (IkAextEBw⇤B � qk))T + (�kUB � �k

LB)T Anm)e, (6.5a)

subject to Ainte = 0, (6.5b)

1T e 1, (6.5c)e � 0. (6.5d)

In the new MP, the former constraint (6.2b) has now been complemented with(6.4c)-(6.4f), where the unmeasured rates are specified by the two vectors qUB

nm ,and qLB

nm, and Anm are the rows of the unmeasured extracellular metabolites in thestoichiometric matrix. The MP objective (6.2a) has been complemented with apenalty function where MUB and MLB are to be set su�ciently large to satisfy thenew bounds. Finally, the new constraints on the bounds have been introduced intothe SP objective (6.3a) via dual variables �k

UB and �kLB.

2 Based on the results in the paper, measurement errors appeared to have little e↵ect on whatEFMs that were identified to fit the data set.

103

Page 122: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

6. Contribution of Paper II: A column generation-based method for pathwayidentification in complex metabolic networks

6.3 Summary of the contributionTo conclude, the main contribution of Paper II is a novel method based on the CGtechnique that can solve the EFMs-based MFA problem to optimality, even whenthe underlying metabolic reaction network is complex.

While CG is a well-established technique within mathematical optimization(Lübbecke and Desrosiers, 2005), to determine whether CG is applicable to a certainproblem, and how to formulate the MP and SP(s), are not necessarily straightforward.Overcoming those challenges can realize the e�ciency and potential time savingso↵ered by the CG technique. Here, the application of the CG technique enabled usto e�ciently solve a complex optimization problem encountered in biotechnologicalapplications.

The presented method will be useful in situations where the goal is to identify asubset of pathways in a metabolic reaction network, i.e. a set of of EFMs, relevantto an experimental data set in terms of optimizing the quality of a data-fitting. Byuse of the CG technique, the user will not be limited to an extensively simplifieddescription of cell metabolism. Meanwhile, the method does not replace an EFMenumeration in a more general sense as it does not provide E, meaning a fullenumeration of the EFMs of a network, and it does neither attempt to maximize thenumber of EFMs in EB.

104

Page 123: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Chapter 7

Contribution of Paper III:Integration of thecolumn-generation-basedpathway identification into thepoly-pathway methodology

The aim in Paper III was to continue the work initiated in Paper I, but now withaccess to the CG-based method presented in Paper II. This chapter begins with thespecification of a suitable network and the identification of EFMs for the model(Section 7.1). Next, kinetic model development is revisited and new features areintroduced, primarily related to the parameter estimation and model evaluation(Section 7.2). Finally, Section 7.3 summarizes the main contributions of the paper.

7.1 Network and identification of EFMs for the modelFirst, reconsider the workflow in Figure 5.3 (p. 87) where the objective was toreach proof-of-concept. It was recognized that the approach would benefit from thepossibility to use a larger and more flexible network as input (2); a user should nothave to sacrifice highly relevant network properties. A potential solution based onthe CG technique was proposed in Paper II.

105

Page 124: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7. Contribution of Paper III: Integration of the column-generation-basedpathway identification into the poly-pathway methodology

Figure 7.1: Metabolic network in Paper III. The metabolites are listed on p. ix. This samemap is found in Figure 2 of Paper III.

This section first covers the integration of CG into the previous workflow,meant to replace the EFM enumeration (3) by an identification of EFM subsets(Section 7.1.1). This is followed by a description of the new and significantly more

106

Page 125: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7.1. Network and identification of EFMs for the model

complex proof-of-concept network N126 (Section 7.1.2). Finally, the result of theEFM identification is briefly discussed (Section 7.1.3).

7.1.1 Integration of CG into the modeling framework

A first task was the integration of CG into the modeling workflow. As noted earlier(p. 102), the CG-based method solves the EFMs-based MFA problem for a singlepseudo-steady state, and does not account for multiple experimental conditions orkinetics. It achieves an identification of an EFM subset that is relevant to a specificexperimental condition in terms of a data-fitting. With Principle 2 of the metabolicmodel (p. 84), that EFM subset could be interpreted as the pathways in use by thecell line when cultured in a specific medium (but with reservations concerning theuniqueness of that particular solution, see p. 101).

7.1. Network and identification of EFMs for the model

complex proof-of-concept network N126 (Section 7.1.2). Finally, the result of theEFM identification is briefly discussed (Section 7.1.3).

7.1.1 Integration of CG into the modeling framework

A first task was the integration of CG into the modeling workflow. As noted earlier(p. 100), the CG-based method solves the EFMs-based MFA problem for a singlepseudo-steady state, and does not account for multiple experimental conditions orkinetics. It achieves an identification of an EFM subset that is relevant to a specificexperimental condition in terms of a data-fitting. With Principle 2 of the metabolicmodel (p. 82), that EFM subset could be interpreted as the pathways in use by thecell line when cultured in a specific medium (but with reservations concerning theuniqueness of that particular solution, p. 99).

(2)Network

Data 1 CGa EB.1, wB.1

Bounds 1

Data 2 CGa EB.2, wB.2

Bounds 2

.

.

....

.

.

.

Data 16 CGa EB.16, wB.16

Bounds 16

Form Eunion

4Macroscopic

reactions

(1)E

xper

imen

tald

ata

(3) EFM identification

EFMsubsets

Figure 7.2: Integration of CG to replace EFM enumeration in the modeling framework. AnEFM subset is identified for each condition. Eunion is the union of those subsets.

The strategy chosen for the development of a single kinetic model is depictedin Figure 7.2. An independent EFM identification was carried out for each exper-imental condition (3). Hence, sixteen EFMs-based MFA problems were solved,

107

Figure 7.2: Integration of CG to replace EFM enumeration in the modeling framework. AnEFM subset is identified for each condition. Eunion is the union of those subsets.

The strategy chosen for the development of a single kinetic model is depictedin Figure 7.2. An independent EFM identification was carried out for each exper-imental condition (3). Hence, sixteen EFMs-based MFA problems were solved,

107

Page 126: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7. Contribution of Paper III: Integration of the column-generation-basedpathway identification into the poly-pathway methodology

yielding sixteen EFM subsets, one for each pseudo-steady state in the experiment.The sixteen EFM subsets were then combined into one, by forming their union,

Eunion = E1 [ E2 [ E3 [ ... [ E16. (7.1)

Here, Eunion

(i) contains only unique EFMs,

(ii) is a subset of all the EFMs in E,

(iii) and can solve each of the sixteen EFMs-based MFA problems to optimality,achieving the same minimized objective function value as would be obtainedif all the EFMs were known beforehand.

Next, Eunion was translated into macroscopic reactions, which corresponds to step 4in Figure 7.2.

7.1.2 Metabolic network N126

The integration of CG into the workflow enabled an increase in the complexity ofthe input network (2). The new proof-of-concept network, in this thesis referred toas N126, was an adapted version of the detailed network by Zamorano et al. (2010).The following are the essential properties of N126.

• 126 reactions, 89 intracellular metabolites, and 29 extracellular metabolites.

• 74 reversible reaction fluxes (58 %).

• Fifteen metabolic subsystems, including e.g. the PPP, EAA catabolism, theUrea cycle, nucleotide synthesis, and lipid synthesis.

• Further compartmentalization, i.e., a mitochondrial subspace and severaltransport reactions.

• Biomass synthesis from intracellular macro-molecules.

• mAb synthesis

• All necessary extracellular metabolites have intracellular counterparts.

The N126 network map is shown in Figure 7.1 (p. 106). A comparison of N126against N30 in Paper I (Figure 5.4) indicates that the di↵erence in complexity levelis apparent, an observation that can be confirmed by inspection of the networkproperties (see Table 3.4, p. 47).

108

Page 127: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7.2. Kinetic model development and evaluation

As could be expected, the EFMs of network N126 could not be enumeratedusing the Metatool algorithm, indicating a high number of possible EFMs. Again,and as a comparison, a model of E. coli with ⇡100 reactions was estimated to have⇡272 million EFMs (Jungreuthmayer and Zanghellini, 2012; Zanghellini et al.,2013).

7.1.3 Identification of the EFMs

Here follows a short description of the EFM identification in Paper III.

• The number of EFMs in the sixteen subsets ranged from 22–25 EFMs perexperimental condition (Table 2, Paper III), which was similar to the numberobtained in the Paper II case-study for the N2 network of similar size.

• Eunion contained 125 unique EFMs, just a third of the 379 EFMs obtainedthrough enumeration of the EFMs in the much simpler N30 network.

• The EFM subsets overlapped, meaning that in some cases the same EFMcould be found in more than one of the subsets (see Figure 6, Paper III).

Before moving on to the kinetic model development, it can be noted that asequence of EFM identifications can be carried out either independently for eachpseudo-steady state, as was done in the present case, or by using EFM outputs fromone or several previous runs as input for the next one. In preliminary test runs,an independent run resulted in fewer overlaps, and this approach was opted for inPaper III to get a more diverse set of EFMs in Eunion.

7.2 Kinetic model development and evaluationThis section revisits the kinetic model development to point out some of the newfeatures introduced in Paper III, i.e. weighting factors, a model reduction, and across-validation with model selection. An overview is provided in Table 7.1.

7.2.1 Weighting factors

The first feature was the introduction of weighting factors into the parameter es-timation to improve the fit for rates deviating from the common patterns in data(Section 5.3 in Paper III). For specific metabolites, deviation from the control culturetypically occurred in a small number of the conditions. The associated data points

109

Page 128: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7. Contribution of Paper III: Integration of the column-generation-basedpathway identification into the poly-pathway methodology

Table 7.1: Features added to the kinetic model development in Paper III.

Observation New feature What it doesThe data set encompass sixteenconditions, but unusual behav-ior tends to be less well cap-tured in LSQ.

Weighting factors Assigns higher (or lesser)weight to selected data pointsto promote simulation of un-usual behavior.

A flexible model structureyields a large initial model withmany equations.

Model reduction Identifies the model equationscontributing significantly to thesimulations, and forms a re-duced model.

The model is evaluated againstthe same data that was used totrain it. Risk of overfitting.

Cross-validation Assesses the ability of themodel to predict unseen testdata. Selects a best model interms of predictive capacity.

were therefore in minority. For example, if one condition deviated from a commonpattern displayed in the other fifteen conditions, the data points associated with itconstituted only ⇡ 6.25 % of the data on that metabolite. However, in a standardleast-squares fitting, the optimization problem does not di↵erentiate between theexperimental conditions. It is simply guided by its objective which is to minimizethe error between the model and the data as a whole. The principle is illustratedin Figure 7.3. Initially, a first model may fit the common patterns in the data well,but miss the unusual behavior of the cell line. The fact that the unusual behavior isimportant has not been conveyed.

By introducing weighting factors, a better model can be obtained. The datapoints associated with the unusual behavior can be assigned higher weight, so thatthe new model captures both the common pattern and the deviations reasonablywell. In the context of the present work, the latter model is a better model in termsof capturing diverse behavior.

7.2.2 Model reduction

The second feature to be introduced was a simple model reduction step. In Paper I,the presented proof-of-concept model encompassed the entire equation pool. Infact, a large fraction of the values in wmax were zero or close to zero, so only asmall fraction of the equations contributed significantly to the simulations. The

110

Page 129: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7.2. Kinetic model development and evaluation

LSQ

LSQ with weighting factors

Figure 7.3: A conceptual illustration of the weighting factors. The first model (top) fitsthe common pattern of behavior well, but misses the unusual behavior deviating from thatpattern. Assigning higher weights to the data points associated with the unusual behavioryields a second model (bottom), better able to capture also the diverse behavior.

same observation was made in Paper III. This provided an opportunity to reduce themodel in size.

The new model reduction step entailed that all equations fitted by a wmax lowerthan a certain cut-o↵ value were removed from the model (Figure 7.4). Thisyielded a significantly smaller model. In fact, keeping approximately 5–10 % of theequations in the equation pool gave a similar fitting error as before the reduction(Figure 7.4B).

7.2.3 Cross-validation and model selection

The final feature was the cross-validation carried out to assess overfitting, and toselect a final proof-of-concept model for the paper. Cross-validation challenges amodel to predict unseen data, which can reveal strengths and weaknesses of themodel itself, as well as the methodology used to generate it. The insights gainedcan then inspire future research and development (Cawley and Talbot, 2010). In across-validation, a data set is divided into a training set and a test set. The trainingset is used to train a model, and that model is then used to predict the test set (Hastieet al., 2009).

111

Page 130: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7. Contribution of Paper III: Integration of the column-generation-basedpathway identification into the poly-pathway methodology

Equation poolInitialmodel

Reducedmodel

Estimate Sort Reduce

CF

1A1B2A2B2C

34

5A5B

67A7B7C

3

7D8A8B

910

Pathways

123456789

10

Kinetics

2B6

1A5B7B2A

45A7A

32B

61A5B7B2A

4

7B8A

1 2 3 4

109

1B8B7D7C

(A)

(B)

Figure 7.4: Conceptual illustration of data-driven selection of the kinetic functions, nowwith model reduction. (A): As before, a pool of kinetic equations is formulated at step 1.After the linear parameter estimation at step 2, the equations are sorted at step 3. (B): Acut-o↵ value wcut for the model parameter wmax is determined to separate equations that willbe kept, or discarded, respectively. For details, see Figure 7 in Paper III. (A) continued: Atstep 4, the equations associated with parameter values above the cut-o↵ value are used todefine a reduced model.

112

Page 131: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7.2. Kinetic model development and evaluation

the parameter estimation (i.e., the weighting matrix 7k is a diagonalmatrix in which the entries are the weights given to each row in (10)).The fluxes with higher information content are attributed a higherweight as commonly used to select the weights during an iterativemodelling exercise (Montgomery et al., 2015) so that the model fit isimproved. This strategy is used to improve the fit for particularvariations in some experimental conditions that are not well captured

in the preliminary estimations. Furthermore, the data with a largeruncertainty are attributed a lower weight, e.g. lysine uptake. Theresulting model is an initial and large model (Step 1), LargeMinitial,including the macro-reactions (or EFMs) and the kinetics. Thesimulated fluxes generated with this model are compared to themeasured fluxes in Supplementary material Figs. S5–S7 showing agood agreement of the model to the data.

Fig. 4. Determination of the macro-reactions and kinetics - Step 1. The macro-reactions of the metabolic reaction network are created by an EFM approach: given the metabolicnetwork, the experimental data and the bounds, the Eunion is generated using the CG algorithm (see Fig. 3). One or several potential kinetic equations with saturation and/or inhibition

effects are attributed for each EFM in Eunion, noted as ‘All the potential kinetics equations associated to each macro-reaction’. This generates a large potential model, LargeM, composed

of the macro-reactions and several kinetics alternative per macro-reaction. The potential kinetic equations vary in the effect of the metabolites. The saturation parameters Ks of the

kinetics are taken from the literature while the inhibition parameters Kp and Kr are obtained by fitting the fluxes estimated by the model to the experimental data in LargeM.

Fig. 5. Identification of the reduced model - Step 2. A cross-validation approach is applied to identify the final model. The data are randomly distributed in a training data set and atesting data set. For each training data set d, the macro-reaction network defined by Eunion and the kinetics of Step 1 are used to determine a large model LargeMd. The LargeMd is

reduced into RedMd. RedMd is then used on the testing data set to simulate the flux rates and the error between the simulated and measured data is computed. From all the repetitionexercises, the RedMd providing the smallest simulation error for the testing data is selected as final model RedMfinal.

E. Hagrot et al. Metabolic Engineering Communications 8 (2018) e00083

7

Figure 7.5: Overview of the cross-validation and model selection in Paper III. This samefigure is Figure 5 in the paper.

Cross-validation can be applied to examine the performance of the model outsidean experimental range, see e.g. Grosfils et al. (2007), or to assess overfitting issues(Almquist et al., 2014). Overfitting can be a problem in large models; there is a riskthat the model reproduces the variation between particular data points, rather thanthe variations resulting from the actual phenomena under study.

In a k-fold cross-validation, the data is divided into k folds. The cross-validationis repeated k times, such that each fold has been the test set once. This type ofcross-validation can be applied in model selection (Cawley and Talbot, 2010); itgenerates a set of alternative models from which a best model can be selected basedon specified criteria. In Paper III, a 4-fold cross-validation was applied and repeated10 times.

As depicted in Figure 7.5, the validation began from Eunion, the entire kineticequation pool, and the associated parameters Ks, Kp and Kr. The data were splitvia a random assignment into four folds of approximately equal size, and in astratified manner such that in each fold all sixteen scenarios were fully represented.The procedure included a model reduction step. The results did not indicate any

113

Page 132: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7. Contribution of Paper III: Integration of the column-generation-basedpathway identification into the poly-pathway methodology

extensive overfitting issues. In general, the output predicted for the test data weresimilar to the output simulated using the reference model (Figures 8–10, Paper III).

Next, the best model was the one with the best predictive capacity (see e.g.the MaxOut ideal, p. 38), i.e. the model out of all the candidates with the bestdata-fitting performance in terms of smallest error between model and testing data(Figure 7.5). As a result of the model reduction and model selection, the selectedmodel, RedMfinal, consisted of only 65 of the initial 125 macroscopic reactions (seeSection 5.5, Paper III).

7.3 Summary of the contributionPaper III continued to explore the relevant question of how to generate a single cellculture model with ability to simulate diverse behavior. The main contribution ofPaper III was the integration of the CG-based approach from Paper II into the PaperI workflow, and the fact that this integration allowed the use of significantly morecomplex and flexible network in kinetic modeling.

While the integration of CG gave an improved data-fitting, the most strategicway to carry out multiple EFM subset identifications could be examined in futurework. The option to use EFM subsets as input has been implemented into thealgorithm. Hence, di↵erent run orders and combinations could be carried outsystematically to examine alternative solutions, and perhaps to determine a core setof EFMs for a model.

In Paper III, the network/EFM identification and the kinetic model developmentare still treated separately, as the CG approach can only handle individual pseudo-steady states. A simultaneous EFM identification and kinetic parameter estimationis an interesting but challenging prospect. If realized, the EFM identification wouldaccount for the kinetics that unify the diverse behavior, it would thus strive toidentify the EFMs that encompass the data as a whole. As a starting point, aheuristic approach has been suggested in the thesis by Oddsdóttir (2015).

In addition to the integration of the CG technique, Paper III also introducedseveral new features for the kinetic model development. Firstly, the weightingfactors were introduced to assign higher or lower weight for selected data points.This technique will be useful when the aim of the model is to capture diversebehavior, and especially unusual behavior which can be of high interest from theperspective of biology and process optimization. The weighting factors will also beuseful to assign lower weight to uncertain or low-quality data.

114

Page 133: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

7.3. Summary of the contribution

Secondly, a model reduction was a natural step to take, given that a majority ofthe kinetic equations suggested remained "unused" after the parameter estimation.In Paper III, the generation of a large initial model, LargeM, is just an intermediatestep within the workflow. This is in line with the flexible kinetics strategy; severalkinetic equations are suggested, but which of those that will be retained in the modelis determined by the data.

Model reduction is motivated by guiding ideals such as simplicity or 1-causal,and can result from a practice of minimalist idealization (Chapter 2, p. 37). Inthe present work, the large model with many parameters is rather di�cult to graspand cumbersome to handle. A reduced version of the model simplifies and speedsup subsequent calculations, and pinpoints the reactions and the kinetics that areresponsible for capturing the data in a particular simulation; those are, "the factorsthat make a di↵erence".

Finally, cross-validation and model selection were introduced. In the future,these could be integrated into the workflow as automated procedures. In addition, ifa future increase in experimental throughput can be assumed, a larger portion of theexperimental data could be withheld as test data prior to any model development.Cross-validation would then be fully integrated into the method, and models couldbe updated as new data come in.

115

Page 134: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work
Page 135: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Chapter 8

Contribution of Paper IV:Pathway identification in agenome-scale CHO metabolicnetwork using the columngeneration-based approach

The aim of the study in Paper IV was to challenge the algorithm introduced in PaperII to identify pathways in a highly complex network. For this purpose, a networkwas derived from the CHO cell genome-scale model by Selvarasu et al. (2012) andsubjected to both conventional flux analysis and pathway analysis using data of thecontrol culture from the experiment described in Chapter 4. Section 8.1 presentsthe genome-scale network and the adaptations that were made for the presentwork. Section 8.2 follows with a description of the pathway analysis, and outlinessuggestions for how to extract information from the results. Finally, Section 8.3summarizes the main contributions of the paper.

8.1 The genome-scale network modelThe genome-scale metabolic model by Selvarasu et al. (2012) was publicly availablewhen the study was initiated. As mentioned in Chapter 3, this model is based ona previous mouse model by the same research team (Selvarasu et al., 2010). This

117

Page 136: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

8. Contribution of Paper IV: Pathway identification in a genome-scale CHOmetabolic network using the column generation-based approach

section begins by outlining some of the properties of the initial network model(Section 8.1.1). Next follows a description of, and reasoning behind, the adaptationscarried out to prepare the model for the pathway analysis (Section 8.1.2).

8.1.1 Properties of the initial network model

The following properties of the initial network model by Selvarasu et al. (2012) areindicative of its complexity:

• 1,545 reactions and 1,351 metabolites, whereof 218 were extracellularmetabolites.

• A division into three compartments: extracellular space, cytosol, andmitochondrion.

• Thirteen metabolic subsystems with reactions further categorized into69 metabolic pathways.

• Biomass synthesis from a range of intracellular components, e.g. glycogen,various lipids and cholesterol, co-factors, amino acids, etc.

• 679 reversible reaction fluxes.

For a comparison with the other networks in this thesis revisit Chapter 3 (p. 47).During the preliminary work, the model was analyzed with conventional FBA

using the COBRA toolbox functions (Schellenberger et al., 2011). This revealedsome less obvious properties of the network. Firstly, there were many dead ends;this occurs when reactions produce intracellular metabolites not used anywhereelse in the network (Santos et al., 2011). Secondly, many futile cycles and parallelreactions appeared during the flux analyses. Fluxes in the futile cycle fluxes aredi�cult to determine by flux analysis, as are the fluxes for parallel reactions thatcarry out identical or very similar metabolite conversions (Martens, 2007; Ahn andAntoniewicz, 2012; Pereira et al., 2016).

8.1.2 Adaptations

Before the flux analyses and CG-method could be applied to the network model,several adaptations were required (Section 3.1 and 3.2, Paper IV). Those adaptationsinvolved the addition or removal of certain reactions and metabolites, changes inthe reversibility/irreversibility of certain reactions or their stoichiometry (i.e. inthe IgG synthesis). These adaptations were made considering e.g. the availability

118

Page 137: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

8.2. Pathway analysis at genome-scale

of components in the medium, and amino acid auxotrophies recently reported forCHO-K1 cells (Hefzi et al., 2016),

The downloaded model came with bounds on the exchange reaction fluxes.Logically, the bounds on exchange fluxes that had been experimentally determinedin the study by Selvarasu et al. (2012) were specified according to their data.Bounds for the unmeasured exchange fluxes had also been assumed. Nevertheless,all of those bounds had to be adapted to the present situation and were set basedon reasoning, and via a trial-and-error approach in which both FBA and EFMidentification were attempted.

An important step involved the removal of all the dead ends. The concept ofEFMs builds on the assumption of a pseudo-steady state (p. 53). Hence, intra-cellular metabolites should never deplete or accumulate within the intracellularcompartments, but must always be consumed and produced at the same overall netrate. However, due to the dead ends, some intracellular metabolites appeared assubstrates and/or products in the macroscopic reactions.

To prevent dead end metabolites from entering the macroscopic reactions, alldead end reactions and metabolites were identified and removed from the model.This led to a significant reduction in the model size: the reduced model adapted forpathway analysis consisted of 994 reactions (previously 1,545) and 700 metabolites(previously 1,351). As an example, the Porphyrin and Chlorophyll Metabolismmetabolic pathway was completely removed. While it is true that the pathway playsan important function in heme biosynthesis and catabolism in mammalian cells(Bohler and Willighagen, 2018), that function did not appear to be represented inthe network model, and the metabolites could not be properly connected with theother parts of the network.

8.2 Pathway analysis at genome-scaleThis section is focused on the pathway analysis and the results obtained. Section 8.2.1describes the identification of the EFMs, and insights gained concerning the di�culttask of assuming a large number of bounds to obtain realistic flux distributions.Section 8.2.2 provides suggestions for how to extract information from resultsobtained from the pathway analysis.

8.2.1 EFM identification via column generation

A central question prior to the study was if the CG technique would be able toidentify EFMs in a genome-scale network. After adaptation of the network and

119

Page 138: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

8. Contribution of Paper IV: Pathway identification in a genome-scale CHOmetabolic network using the column generation-based approach

setting bounds as guided FBA, the CG-based algorithm was able to identify 28EFMs with a run time of approximately 3 seconds, a result indicating that theCG-based algorithm can be applied at genome-scale, and that the computation caneven be time e�cient on a single laptop computer.

While it was possible to generate the EFMs, an issue that should be dealt with infuture work was also identified. Genome-scale modeling sets out to achieve a nearlycomplete representation of the biological system, yet a lack of information rendersthe system of equations underdetermined. If valid assumptions can be defined, theresults are likely more precise in terms of more narrow flux intervals. While this istrue, the addition of such constraints can make the problem infeasible. The valuesthemselves may be wrong, infeasible together, or with the data and that particularnetwork.

In the present case, the problem to solve was certainly underdetermined giventhe small set of culture variables that had been measured experimentally. Theanalysis therefore depended to a large extent on the assumed bounds on the exchangereactions. Certain values on those bounds made the FBA problem infeasible. Inthe pathway analysis the algorithm got stuck. Suitable bounds had to be found viatrial-and-error. A way around this problem was to lower the parameter values of theMP penalty function in the MP objective (6.4a), p. 103.

8.2.2 Extracting information from the EFMs

As outlined in Table 3.5 (p. 58), pathway analysis and specifically EFM analysiscan provide valuable information about the structure of metabolic networks. Thecontribution of the network reactions in each pathway are specified directly ineach vector el, and Amac defines the macroscopic reactions of each pathway. Themacroscopic reactions obtained in Paper IV are presented in Table 2, 3, and 4 of thepaper. In Table 8.1 below follows suggestions for how to extract information froman EFM subset, with referral to the heatmaps presented in Paper IV.

8.3 Summary of the contributionTo conclude, the main contribution of Paper IV is the application of the CG-basedmethod introduced in Paper II to a network at genome-scale, showing that it ispossible to obtain an EFM subset via a data-driven pathway identification using theCG technique.

At genome-scale, new types of challenges arise, e.g. dead ends, a huge set ofunmeasured fluxes, and many futile cycles and parallel reactions. To deal with these

120

Page 139: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

8.3. Summary of the contribution

Table 8.1: Extracting information from the EFMs and the macroscopic reactions.

Explore... Solution...the participation of theextracellular metabolites inthe macroscopic reactions.

A heatmap of the macroscopic matrix Amacdisplays the non-zero stoichiometric coe�cientsin a grid where each row corresponds to an extra-cellular substrate or product, and each column toa macroscopic reaction/EFM (Figure 3, Paper IV).In Amac the substrates have a negative element, andthe products a positive element

...which subsystems in themetabolism that are involvedin the macroscopic conver-sions, and the number of par-ticipating reactions in eachof them.

For each el, identify all the positions for the reac-tions belonging to a specific subsystem, and countthe non-zero elements. This generates a matrixfor the subsystem usage in which each row corre-sponds to a subsystem in the network, and eachcolumn to a macroscopic reaction/EFM (Figure 4,Paper IV).

...which network pathwaysin each subsystem in themetabolism that are involvedin the macroscopic conver-sions, and the number of par-ticipating reactions in eachof them.

For each el, identify all the positions for the reac-tions belonging to a specific network pathway, andcount the non-zero elements. This generates a ma-trix for the network pathway usage in which eachrow corresponds to a network pathway, and eachcolumn to a macroscopic reaction/EFM (Figure 5,Paper IV).

issues require that many assumptions are made; there is a risk that the problembecomes infeasible. A relaxed form of CG, as is available for FBA in the COBRAtoolbox, may help to tackle this issue.

At the initiation of this thesis work, genome-scale modeling of CHO cells wasless developed. Since then, major progress has been achieved in this area, anddetailed models of popular CHO cell lines are available for download (Hefzi et al.,2016). Still, the experimental data set in this thesis is very small considering thehigh complexity of those models.

121

Page 140: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work
Page 141: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Chapter 9

Conclusions and futureperspectives

The demand for biopharmaceuticals will likely persist in the future, as will the im-portance of CHO and other mammalian cell lines for their production. Mathematicalmodeling has the potential to dramatically accelerate the research in this area. Inthis thesis, the question of how to generate such models has been examined, withthe starting point in the macroscopic kinetic modeling of a mAb-producing CHOcell line.

The methodology was built on previous work within the field: a first step ofidentifying all the possible pathways through the cell metabolism via conventionalEFM enumeration; and a second step of modeling the kinetics of the pathway fluxesusing generalized Michaelis-Menten-type equations. Previous models of this typehave targeted the simulation of phases during batch cultures, but in the present workthe problem to solve was di↵erent. The aim was to develop a flexible modelingframework with potential for process optimization. Given this long-term goal, acentral requisite was the ability of one single model to capture diverse behavior of acell line in response to variations in the culture conditions.

To obtain data for model development, parallel pseudo-perfusion cell cultureswere carried out, and subjected to di↵erences in the amino acid availability. The ex-periment was purposefully designed to trigger activation of alternative and compen-satory pathways within the metabolism, and involved both doublings and completeomissions of selected single amino acids. While this type of experiment is relevantto biopharmaceutical process development, it has not been a usual target for kineticmodels.

123

Page 142: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Conclusions and future perspectives

In Paper I, a modeling concept based on three main principles was put forward(p. 84): the cells have access to multiple pathways; their observed macroscopicbehavior is a result of combined pathway activities; and those activities are regulatedby the cells in response to environmental changes. Starting from a typical workflowfor macroscopic kinetic modeling, proof-of-concept was achieved after severaliterations and further development.

An important insight in Paper I was that modeling diverse behavior and a flexiblemetabolism call for a flexible model structure. Such flexibility can be achieved bothin the structure of the network, and in the generalized kinetic equations. In addition,there is an important relation between the two; the kinetic functions are determinedautomatically from the macroscopic reactions, unless otherwise instructed. Thistends to lead to a "combinatory explosion", di�cult to handle.

Another important insight in Paper I was that due to the conventional EFMenumeration step, only simple networks could be used to generate models. Thisled to the development of a novel algorithm based on the CG technique (Paper II),which could be integrated into the kinetic modeling framework, and enabled theuse of a more detailed and complex metabolic network (Paper III). In addition, thealgorithm was applied as a stand-alone tool for the identification of a relevant EFMsubset in a genome-scale network (Paper IV).

How to formulate the kinetics in stoichiometric models is subject to research;this involves both the structure of the kinetic equations and the identification oftheir parameters. The present work arrived at a flexible generalized equation. Whilemultiple kinetic equations were formulated, based on the fitting to experimentaldata and steps of model reduction and a cross-validation exercise, only a subsetof the initial equations ended up in a final reduced model (Paper III). The manualtesting of di↵erent equations was moved, at least partially, into the computationalframework for a more objective selection of the kinetics.

Data collection is an important aspect within the modeling framework. Un-fortunately, only a limited number of conditions could be tested in the parallelcell cultures; the protocol relied largely on manual work and had limited access totechnical equipment and analytical methods. Automated equipment referred to as"robot culture machines" are nowadays in use within the industry. While expensive,these machines can increase the experimental throughput significantly, in particularif automated sampling and liquid handling can be combined with advanced high-throughput analyses. Furthermore, the experimental design was a first approach. Infuture work, methods such as identifiability analysis (Almquist et al., 2014) couldguide the set up of experiments that produce data adequate to identify a model.

Future work could involve learning more about each particular pseudo-steady

124

Page 143: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Conclusions and future perspectives

state in the experimental data. This could mean additional measurements, a morethorough literature review, and flux analyses. While this knowledge would beincorporated into the model, the model development and resulting simulations couldin turn contribute to explanation, i.e. for how the availability of certain amino acidsinfluence the metabolism of the cells. Here the flux maps are useful in order tovisualize and compare the intracellular flux distributions underlying a model, andalso serve as a validation of the model against what is biologically reasonable.

Further automation is possible within the kinetic modeling workflow. Adjust-ments of the network properties, and the addition and evaluation of kinetic equationscould be performed automatically within a program, given that the proper instruc-tions are provided. A major achievement would be the development of theory for,and the subsequent implementation of, a simultaneous identification of pathwaysand kinetic parameters, as was discussed in Chapter 7.

Parameter estimation in the complex and non-linear systems found in biologycan be a challenge, and is often prohibitive. Nevertheless, it provides opportunitiesfor research, e.g. to formulate and simplify problems, and to develop e�cientsolvers and strategies for estimating parameter values. Such e↵orts are currentlyundertaken under the lead of the Department of Automatic Control at KTH withinthe Center for Advanced Bioprocessing by Continuous Process (AdBIOPRO), andin collaboration with the Cell Technology Group (CETEG) at KTH.

The kinetic model methodology and the column-generation-based method aregeneral, and could with appropriate adaptation be applied to other organisms, othermedia, other cultivation modes and parameters. The batch- and fed-batch cultivationmodes could be of interest, as these are widely applied in biopharmaceutical pro-duction. Ideally, a single poly-pathway model would be developed to simulate theentire duration of such cultivations, rather than through a combination of multiplemodels.

Furthermore, physical parameters such as temperature (Nolan and Lee, 2011)or pH (Hagrot, 2011) were outside the scope of the work, as was the glycosylationprofile of the biopharmaceutical product (Jedrzejewski et al., 2014). The modelingof the resulting product glycoform as a function of the medium composition andother process parameters is of great interest to the biopharmaceutical industry(Huang et al., 2017); this has been a research topic in modeling studies (Kontoravdiet al., 2007; Jedrzejewski et al., 2014).

Genome-scale networks are attractive for medium design applications. Thesmall number of metabolites included in most kinetic models does not reflect thecomplex media used in mammalian cell cultures. Genome-scale networks on theother hand may provide additional metabolites, reactions and pathways for the

125

Page 144: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Conclusions and future perspectives

processing of many common medium components. Via pathway analysis, eachmedium component and its involvement in e.g. biomass synthesis can be traced.The CG-based algorithm could prove useful for this type of application. A next stepcould be the development of kinetic models based on those identified pathways tostudy, and simulate, medium component utilization. Unfortunately, the functions ofall the common cell culture medium components are not fully known, and/or arenot yet expressed mathematically in the available network models.

A good help in the examination of EFMs-based pathways in the genome-scalenetworks would be visualizations in a network map in which each participatingreaction would be highlighted. The automatic generation of network maps anddevelopment of the associated software are very interesting topics (Rohn et al.,2012; Copeland et al., 2012; King et al., 2016). The use of consensus models,identifiers, and general standardization are bound to make such visualizations muchmore e�cient (Gerstl et al., 2017).

When no or little experimental data is available, flux analysis problems tendto be underdetermined with many feasible solutions. This is critical for very largereaction networks. As demonstrated in Paper IV, while the CG-based algorithmcould deliver a solution which was optimal in a mathematical sense, biologicalrelevance can generally not be guaranteed. Such relevance need to be enforcedby formulating and implementing biological assumptions as constraints, and/or byobtaining additional data, which should be considered in future applications of theapproach.

It can be emphasized that throughout the work, calculations were carried outlocally on a single personal computer, that is, without access to e.g. parallelcomputing (Terzer and Stelling, 2008). The acceptable time frame for an EFMidentification was in the range of seconds, rather than hours or a full working day.On one hand, this favors the general usefulness of the developed algorithm. On theother hand, one might expect that pathway analysis at genome-scale guided by theintegration of a high number of constraints and various omics’ data would requirelong run times, and/or depend on extensive computational power.

To conclude, we can imagine a future in which high-throughput data generation,knowledge about biological systems, and the development of useful modelingapproaches, are combined to accelerate the development of biopharmaceuticalprocesses. Biopharmaceuticals are of considerable economic value to the industry.But even more important is the ability of these complex biological molecules tocombat the health issues encountered by people all over the world. Hopefully, inthe future, both previous and new biopharmaceutical products can be e�cientlyproduced, and be accessible and a↵ordable to everyone that needs them.

126

Page 145: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

ReferencesWoo Suk Ahn and Maciek R. Antoniewicz. Towards dynamic metabolic flux analysis in

CHO cell cultures. Biotechnol. J., 7(1):61–74, 2012.

Woo Suk Ahn and Maciek R. Antoniewicz. Parallel labeling experiments with [1,2-(13)C]glucose and [U-(13)C]glutamine provide new insights into CHO cell metabolism.Metab. Eng., 15:34–47, 2013.

Joachim Almquist, Marija Cvijovic, Vassily Hatzimanikatis, Jens Nielsen, and MatsJirstrand. Kinetic models in industrial biotechnology — Improving cell factory per-formance. Metab. Eng., 24:38–60, 2014.

Hal Alper, Kohei Miyaoku, and Gregory Stephanopoulos. Construction of lycopene-overproducing E. coli strains by combining systematic and combinatorial gene knockouttargets. Nat. Biotechnol., 23(5):612–616, 2005.

Claudia Altamirano, Andres Illanes, Silvana Becerra, Jordi Joan Cairó, and Francesc Gòdia.Considerations on the lactate consumption by CHO cells in the presence of galactose. J.Biotechnol., 125(4):547–56, 2006.

Claudia Altamirano, Julio Berrios, Mauricio Vergara, and Silvana Becerra. Advances inimproving mammalian cells metabolism for recombinant protein production. Electron. J.Biotechnol., 16(3), 2013.

P. L. Altman and D. S. Dittmer. Respiration and circulation. Federation of AmericanSocieties of Experimental Biology, MD., 1971.

Jasbir S. Arora. Duality in Linear Programming. In Introduction to Optimum Design,chapter 9.3, pages 399–414. Academic Press, Boston, fourth edition, 2017.

John G. Aunins and Hans-Jürgen Henzler. Aeration in Cell Culture Bioreactors. In H.-J.Rehm and G. Reed, editors, Biotechnology: Bioprocessing, Volume 3, chapter 11, pages219–281. VCH Verlagsgesellschaft mbH, second edition, 2008.

James E. Bailey. Mathematical modeling and analysis in biochemical engineering: Pastaccomplishments and future opportunities. Biotechnol. Prog., 14(1):8–20, 1998.

J. Barthelmes, C. Ebeling, A. Chang, I. Schomburg, and D. Schomburg. BRENDA,AMENDA and FRENDA: the enzyme information system in 2007. Nucleic AcidsRes., 35(Database issue):D511–D514, 2007.

Bassem Ben Yahia, Laetitia Malphettes, and Elmar Heinzle. Macroscopic modeling ofmammalian cell growth and metabolism. Appl. Microbiol. Biot., 99(17):7009–7024,2015.

127

Page 146: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

L. R. Bertolini, H. Meade, C. R. Lazzarotto, L. T. Martins, K. C. Tavares, M. Bertolini,and J. D. Murray. The transgenic animal platform for biopharmaceutical production.Transgenic Res., 25(3):329–343, 2016.

Rahul Bhambure, Kaushal Kumar, and Anurag S. Rathore. High-throughput processdevelopment for biopharmaceutical drug substances. Trends Biotechnol., 29(3):127–135,2011.

Theodora A. Bibila and David K. Robinson. In pursuit of the optimal fed-batch process formonoclonal antibody production. Biotechnol. Prog., 11(1):1–13, 1995.

Anwesha Bohler and Egon Willighagen. Metabolism of porphyrins (Homo sapiens), 2018.URL https://www.wikipathways.org/index.php/Pathway:WP1852.

Hendrik P. J. Bonarius, Georg Schmid, and Johannes Tramper. Flux analysis of underdeter-mined metabolic networks: the quest for the missing constraints. Trends Biotechnol., 15:308–314, 1997.

Hendrik P. J. Bonarius, Bram Timmerarends, Cornelis D. De Gooijer, and Johannes Tramper.Metabolite-balancing techniques vs. 13C tracer experiments to determine metabolicfluxes in hybridoma cells. Biotechnol. Bioeng., 58(2-3):258–262, 1998.

Henrik P. J. Bonarius, Vassily Hatzimanikatis, Koen P. H. Meesters, Cornelis D. de Gooijer,Georg Schmid, and Johannes Tramper. Metabolic flux analysis of hybridoma cells indi↵erent culture media using mass balances. Biotechnol. Bioeng., 50(3):299–318, 1996.

Scott A. Bradley, Anli Ouyang, Jennifer Purdie, Tim A. Smitka, Tongtong Wang, andAndreas Kaerner. Fermentanomics: Monitoring mammalian cell cultures with NMRspectroscopy. J. Am. Chem. Soc., 132(28):9531–9533, 2010.

Mario Bunge. A General Black Box Theory. Philos. Sci., 30(4):346–358, 1963.

A. Burgener and M. Butler. Medium Development. In Sadettin S. Ozturk and Wei-ShouHu, editors, Cell Culture Technology for Pharmaceutical and Cell-Based Therapies,chapter 3, pages 41–79. CRC Press, Taylor & Francis Group, Boca Raton, FL, firstedition, 2006.

Michael Butler and Maureen Spearman. The choice of mammalian cell host and possibilitiesfor glycosylation engineering. Curr. Opin. Biotechnol., 30:107–112, 2014.

Yihua Cao, Shuichi Kimura, Takayuki Itoi, Kohsuke Honda, Hisao Ohtake, and TakeshiOmasa. Construction of BAC-based physical map and analysis of chromosome rear-rangement in chinese hamster ovary cell lines. Biotechnol. Bioeng., 109(6):1357–1367,2012.

128

Page 147: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Nuno Carinhas, Tiago M. Duarte, Laura C. Barreiro, Manuel J.T. Carrondo, Paula M.Alves, and Ana P. Teixeira. Metabolic signatures of GS-CHO cell clones associatedwith butyrate treatment and culture phase transition. Biotechnol. and Bioeng., 110(12):3244–3257, 2013.

L. M. Carrillo-Cocom, T. Genel-Rey, D. Araíz-Hernández, F. López-Pacheco, J. López-Meza, M. R. Rocha-Pizaña, A. Ramírez-Medrano, and M. M. Alvarez. Amino acidconsumption in naïve and recombinant CHO cell cultures: producers of a monoclonalantibody. Cytotechnology, 67(5):809–820, 2015.

Gavin C. Cawley and Nicola L. Talbot. On Over-fitting in Model Selection and SubsequentSelection Bias in Performance Evaluation. J. Mach. Learn. Res., 11:2079–2107, 2010.

Ning Chen, Mark H. Bennett, and Cleo Kontoravdi. Analysis of Chinese hamster ovary cellmetabolism through a combined computational and experimental approach. Cytotechnol-ogy, 66:945–966, 2014.

Peifeng Chen and Sarah W. Harcum. E↵ects of amino acid additions on ammonium stressedCHO cells. J. Biotechnol., 117:277–86, 2005.

Véronique Chotteau, Ye Zhang, and Marie-Francoise Clincke. Very High Cell Density inPerfusion of CHO Cells by ATF, TFF, Wave Bioreactor, and/or CellTank Technologies —Impact of Cell Density and Applications. In Ganapathy Subramanian, editor, ContinuousProcessing in Pharmaceutical Manufacturing, chapter 13, pages 339–356. Wiley-VCHVerlag GmbH & Co. KGaA., first edition, 2015.

Bruce L. Clarke. Stability of Complex Reaction Networks (1980). John Wiley & Sons, Ltd,2007.

Marie Françoise Clincke, Carin Mölleryd, Ye Zhang, Eva Lindskog, Kieron Walsh, andVéronique Chotteau. Very high density of CHO cells in perfusion by ATF or TFF inWAVE bioreactor(TM): Part I: E↵ect of the cell density on the process. Biotechnol. Prog.,29(3):754–767, 2013.

Peter S. Coleman and Beverly B. Lavietes. Membrane Cholesterol, Tumorigenesis, and theBiochemical Phenotype of Neoplasia. CRC Cr. Rev. Biochem., 11(4):341–393, 1981.

Daniel J. Cook and Jens Nielsen. Genome-scale metabolic models applied to human healthand disease. WIRE Syst. Biol. Med., 9:e1393, 2017.

Wilbert B. Copeland, Bryan A. Bartley, Deepak Chandran, Michal Galdzicki, Kyung H.Kim, Sean C. Sleight, Costas D. Maranas, and Herbert M. Sauro. Computational toolsfor metabolic engineering. Metab. Eng., 14(3):270–280, 2012.

129

Page 148: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Hussain Dahodwala, Mark Nowey, Tatyana Mitina, and Susan T. Sharfstein. E↵ects ofclonal variation on growth, metabolism, and productivity in response to trophic factorstimulation: A study of Chinese hamster ovary cells producing a recombinant monoclonalantibody. Cytotechnology, 64:27–41, 2012.

George B. Dantzig and Philip Wolfe. Decomposition Principle for Linear Programs. Oper.Res., 8(1):101–111, 1960.

Luis F. de Figueiredo, Adam Podhorski, Angel Rubio, Christoph Kaleta, John E. Beasley,Stefan Schuster, and Francisco J. Planes. Computing the shortest elementary flux modesin genome-scale metabolic networks. Bioinformatics, 25(23):3158–3165, 2009.

Maria J. De Jesus, Philippe Girard, Michaela Bourgeois, Gwendoline Baumgartner, BarbaraJacko, Hanspeter Amstutz, and Florian M. Wurm. TubeSpin satellites: a fast trackapproach for process development with animal cells using shaking technology. Biochem.Eng. J., 17:217–223, 2004.

Rahul Deshpande, Tae Hoon Yang, and Elmar Heinzle. Towards a metabolic and iso-topic steady state in CHO batch cultures for reliable isotope-based metabolic profiling.Biotechnol. J., 4:247–263, 2009.

Jacques Desrosiers, François Soumis, and Martin Desrochers. Routing with time windowsby column generation. Networks, 14(4):545–565, 1984.

V. M. Dezengotita, R. Kimura, and W. M. Miller. E↵ects of CO2 and osmolality on hy-bridoma cells: growth, metabolism and monoclonal antibody production. Cytotechnology,28:213–227, 1998.

Penny Dorka, Christian Fischer, Hector Budman, and Jeno M. Scharer. Metabolic flux-basedmodeling of mAb production during batch and fed-batch operations. Bioprocess Biosyst.Eng., 32:183–96, 2009.

Jennifer Dumont, Don Euwart, Baisong Mei, Scott Estes, and Rashmi Kshirsagar. Humancell lines for biopharmaceutical manufacturing: history, status, and future perspectives.Crit. Rev. Biotechnol., 36(6):1110–1122, 2016.

Dawn M. Ecker, Susan Dana Jones, and Howard L. Levine. The therapeutic monoclonalantibody market. mAbs, 7(1):9–14, 2015.

J. S. Edwards and B. Ø. Palsson. The Escherichia coli MG1655 in silico metabolic genotype:Its definition, characteristics, and capabilities. PNAS, 97(10):5528–5533, 2000.

Angela Faustino Jozala, Danilo Costa Geraldes, Louise Lacalendola Tundisi, Valker deAraújo Feitosa, Carlos Alexandre Breyes, Samuel Leite Cardoso, Priscila Gava Mazzola,Laura de Oliveira-Nascimento, Carlota de Oliveira Rangel-Yagui, Pérola de Oliveira

130

Page 149: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Magalhães, Marcos Antonio de Oliveira, and Adalberto Pessoa Jr. Biopharmaceuticalsfrom microorganisms: from production to purification. Braz. J. Microbiol., 47:51–63,2016.

Julia Feichtinger, Inmaculada Hernández, Christoph Fischer, Michael Hanscho, NorbertAuer, Matthias Hackl, Vaibhav Jadhav, Martina Baumann, Peter M. Krempl, ChristianSchmidl, Matthias Farlik, Michael Schuster, Angelika Merkel, Andreas Sommer, Si-mon Heath, Daniel Rico, Christoph Bock, Gerhard G. Thallinger, and Nicole Borth.Comprehensive genome and epigenome characterization of CHO cells in response toevolutionary pressures and over time. Biotechnol. Bioeng., 113(10):2241–2253, 2016.

Adam M. Feist and Bernhard Ø. Palsson. The biomass objective function. Curr. Opin.Biotechnol., 13:344–349, 2010.

Adam M. Feist, Christopher S. Henry, Jennifer L. Reed, Markus Krummenacker, Andrew R.Joyce, Peter D. Karp, Linda J. Broadbelt, Vassily Hatzimanikatis, and Bernhard Ø.Palsson. A genome-scale metabolic reconstruction for Escherichia coli K-12 MG1655that accounts for 1260 ORFs and thermodynamic information. Mol. Syst. Biol., 3:121,2007.

L. R. Ford and D. R. Fulkerson. A Suggested Computation for Maximal Multi-CommodityNetwork Flows. Manage. Sci., 5(1):97–101, 1958.

A. G. Fredrickson, R. D. Megee, and H. M. Tsuchiya. Mathematical Models for Fermenta-tion Processes. Adv. Appl. Microbiol., 13:419–465, 1970.

Komei Fukuda and Alain Prodon. Double Description Method Revisited. In MichelDeza, Reinhardt Euler, and EulerIoannis Manoussakis, editors, 8th Franco-Japanese and4th Franco-Chinese Conference on Combinatorics in Computer Science (1995), Berlin,Heidelberg, 1996. Springer.

Anshu Gambhir, Anna F. Europa, and Wei Shou Hu. Alteration of cellular metabolism byconsecutive fed-batch cultures of mammalian cells. J. Biosci. Bioeng., 87(6):805–810,1999.

Jianying Gao, Volker M. Gorenflo, Jeno M. Scharer, and Hector M. Budman. Dynamicmetabolic modeling for a MAB bioprocess. Biotechnol. Prog., 23(1):168–181, 2007.

Matthias P. Gerstl, Michael Hanscho, David E. Ruckerbauer, Jürgen Zanghellini, and NicoleBorth. CHOmine: An integrated data warehouse for CHO systems biology and modeling.Database, 2017:bax034, 2017.

Atefeh Ghorbaniaghdam, Olivier Henry, and Mario Jolicoeur. A kinetic-metabolic modelbased on cell energetic state: study of CHO cell behavior under Na-butyrate stimulation.Bioprocess Biosyst. Eng., 36:469–487, 2013.

131

Page 150: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Atefeh Ghorbaniaghdam, Jingkui Chen, Olivier Henry, and Mario Jolicoeur. AnalyzingClonal Variation of Monoclonal Antibody-Producing CHO Cell Lines Using an In SilicoMetabolomic Platform. PloS ONE, 9(3):e90832, 2014.

P. C. Gilmore and R. E. Gomory. A Linear Programming Approach to the Cutting-StockProblem. Oper. Res., 9(6):849–859, 1961.

P. C. Gilmore and R. E. Gomory. A Linear Programming Approach to the Cutting StockProblem-Part II. Oper. Res., 11(6):863–888, 1963.

Frances Gódia and Jordi Joan Cairó. Cell Metabolism. In Sadettin S. Ozturk and Wei-ShouHu, editors, Cell Culture Technology for Pharmaceutical and Cell-Based Therapies,chapter 4, pages 81–112. CRC Press Taylor & Francis Group, Boca Raton, FL, firstedition, 2006.

J. M. González-Martínez, A. Folch-Fortuny, F. Llaneras, M. Tortajada, J. Picó, and A. Ferrer.Metabolic flux understanding of Pichia pastoris grown on heterogenous culture media.Chemometr. Intell. Lab., 134:89–99, 2014.

Chetan Goudar, Richard Biener, C. Boisart, Rüdiger Heidemann, James Piret, Albertde Graaf, and Konstantin Konstantinov. Metabolic flux analysis of CHO cells in perfusionculture by metabolite balancing and 2D [13C, 1H] COSY NMR spectroscopy. Metab.Eng., 12:138–149, 2010.

Chetan T. Goudar, Richard Biener, James M. Piret, and Konstantin B. Konstantinov. Meta-bolic Flux Estimation in Mammalian Cell Cultures. In R. Pörtner, editor, Animal CellBiotechnology, volume 24, chapter 14, pages 301–317. Humana Press 2007, secondedition, 2007.

A. Grosfils, A. Vande Wouwer, and Ph. Bogaerts. On a general model structure formacroscopic biological reaction rates. J. Biotechnol., 130:253–264, 2007.

Oliver Hädicke and Ste↵en Klamt. EColiCore2: A reference network model of the centralmetabolism of Escherichia coli and relationships to its genome-scale parent model.Scientific Reports, 7:39647, 2017. doi: 10.1038/srep39647.

Erika Hagrot. Development of a culture system for modeling of pH e↵ects in CHO cells.Master’s thesis, KTH Royal Institute of Technology, 2011.

Erika Hagrot, Hildur Æsa Oddsdóttir, Joan Gonzalez Hosta, Elling W. Jacobsen, andVéronique Chotteau. Poly-pathway model, a novel approach to simulate multiple meta-bolic states by reaction network-based model — Application to amino acid depletion inCHO cell culture. J. Biotechnol., 259:235–247, 2017.

Sarah W. Harcum and Kelvin H. Lee. CHO Cells Can Make More Protein. Cell Systems, 3(5):412–413, 2016.

132

Page 151: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

T. Hastie, R. Tibshirani, and J. Friedman. The elements of statistical learning. Springer-Verlag, New York, seond edition, 2009.

Hooman Hefzi, Kok Siong Ang, Michael Hanscho, Aarash Bordbar, David Ruckerbauer,Meiyappan Lakshmanan, Camila A. Orellana, Deniz Baycin-Hizal, Yingxiang Huang,Daniel Ley, Veronica S. Martinez, Sarantos Kyriakopoulos, Natalia E. Jiménez, Daniel C.Zielinski, Lake Ee Quek, Tune Wul↵, Johnny Arnsdorf, Shangzhong Li, Jae SeongLee, Giuseppe Paglia, Nicolas Loira, Philipp N. Spahn, Lasse E. Pedersen, Jahir M.Gutierrez, Zachary A. King, Anne Mathilde Lund, Harish Nagarajan, Alex Thomas,Alyaa M. Abdel-Haleem, Juergen Zanghellini, Helene F. Kildegaard, Bjørn G. Voldborg,Ziomara P. Gerdtzen, Michael J. Betenbaugh, Bernhard Ø. Palsson, Mikael R. Andersen,Lars K. Nielsen, Nicole Borth, Dong Yup Lee, and Nathan E. Lewis. A ConsensusGenome-scale Reconstruction of Chinese Hamster Ovary Cell Metabolism. Cell Systems,3:434–443, 2016.

Victor Henri. Theorie generale de l’action de quelques diastases. Comptes Rend. Acad. Sci.Paris, 135:916–919, 1902.

Olivier Henry, Ezra Kwok, and James M. Piret. Simpler Noninstrumented Batch and Semi-continuous Cultures Provide Mammalian Cell Kinetic Data Comparable to Continuousand Perfusion Cultures. Biotechnol. Prog., 24:921–931, 2008.

Benjamin J. Hodgson, Christopher N. Taylor, Misti Ushio, J. R. Leigh, Tatiana Kalganova,and Frank Baganz. Intelligent modelling of bioprocesses: A comparison of structuredand unstructured approaches. Bioproc. Biosyst. Eng., 26:353–359, 2004.

Zhuangrong Huang, Dong-Yup Lee, and Seongkyu Yoon. Quantitative intracellular fluxmodeling and applications in biotherapeutic development and production using CHO cellcultures. Biotechnol. Bioeng., 114(12):2717–2728, 2017.

Marija Ivarsson, Heeju Noh, Massimo Morbidelli, and Miroslav Soos. Insights into pH-induced metabolic switch by flux balance analysis. Biotechnol. Prog., 31(2):347–357,2015.

Jae Deog Jang and John P. Barford. An unstructured kinetic model of macromolecularmetabolism in batch and fed-batch cultures of hybridoma cells producing monoclonalantibody. Biochem. Eng. J., 4:153–168, 2000.

Karthik P. Jayapal, Katie F. Wlaschin, and Wei-Shou Hu. Recombinant Protein Therapeuticsfrom CHO Cells — 20 Years and Counting. Chem. Eng. Prog., 103:40–47, 2007.

Philip M. Jedrzejewski, Ioscani Jimenez del Val, Antony Constantinou, Anne Dell, Stuart M.Haslam, Karen M. Polizzi, and Cleo Kontoravdi. Towards controlling the glycoform: Amodel framework linking extracellular metabolites to antibody glycosylation. Int. J. Mol.Sci., 15:4492–4522, 2014.

133

Page 152: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Raphael M. Jungers, Francisca Zamorano, Vincent D. Blondel, Alain Vande Wouwer, andGeorges Bastin. Fast computation of minimal elementary decompositions of metabolicflux vectors. Automatica, 47(6):1255–1259, 2011.

Christian Jungreuthmayer and Jürgen Zanghellini. Designing optimal cell factories: Integerprogramming couples elementary mode analysis with regulation. BMC Syst. Biol., 6:103,2012.

Christian S. Kaas, Yuzhou Fan, Dietmar Weilguny, Claus Kristensen, Helen F. Kildegaard,and Mikael R. Andersen. Toward genome-scale models of the Chinese hamster ovarycells : incentives , status and perspectives. Pharm. Bioprocess., 2(5):437–448, 2014.

Christoph Kaleta, Luís Filipe de Figueiredo, and Stefan Schuster. Can the whole be lessthan the sum of its parts ? Pathway analysis in genome-scale metabolic networks usingelementary flux patterns. Genome Res., 19:1872–1883, 2009.

Minoru Kanehisa and Susumu Goto. KEGG: Kyoto encyclopedia of genes and genomes.Nucleic Acids Res., 28(1):27–30, 2000.

Minoru Kanehisa, Susumu Goto, Yoko Sato, Masayuki Kawashima, Miho Furumichi, andMao Tanabe. Data, information, knowledge and principle: back to metabolism in KEGG.Nucleic Acids Res., 42(Database issue):D199–D205, 2014.

Fa-Ten Kao and Theodore T. Puck. Genetics of somatic mammalian cells. IV. Properties ofChinese hamster cell mutants with respect to the requirement for proline. Genetics, 55:513–524, 1967.

Kazunori Kato. Therapeutic Monoclonal Antibodies for Cancer: The Past, Present, andFuture. Juntendo Medical Journal, 61(6):601–607, 2016.

Ali Khodayari and Costas D. Maranas. A genome-scale Escherichia coli kinetic metabolicmodel k-ecoli457 satisfying flux data for multiple mutant strains. Nat. Commun., 7:13806, 2016.

Ali Khodayari, Ali R. Zomorrodi, James C. Liao, and Costas D. Maranas. A kinetic modelof Escherichia coli core metabolism satisfying multiple sets of mutant flux data. Metab.Eng., 25:50–62, 2014.

Jee Yon Kim, Yeon Gu Kim, and Gyun Min Lee. CHO cells in biotechnology for productionof recombinant proteins: Current state and further potential. Appl. Microbiol. Biotechnol.,93(3):917–930, 2012.

Zachary A. King, Justin Lu, Andreas Dräger, Philip Miller, Stephen Federowicz, Joshua A.Lerman, Ali Ebrahim, Bernhard Ø. Palsson, and Nathan E. Lewis. BiGG Models: Aplatform for integrating, standardizing and sharing genome-scale models. Nucleic AcidsRes., 44(Database issue):D515–D522, 2016.

134

Page 153: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Markus Kirkilionis. Exploration of cellular reaction systems. Brief. Bioinform., 11(1):153–178, 2010.

Ste↵en Klamt and Jörg Stelling. Combinatorial complexity of pathway analysis in metabolicnetworks. Mol. Biol. Rep., 29:233–236, 2002.

Ste↵en Klamt and Jörg Stelling. Two approaches for metabolic pathway analysis? TrendsBiotechnol., 21(2):64–69, 2003.

Ste↵en Klamt, Georg Regensburger, Matthias P. Gerstl, Christian Jungreuthmayer, StefanSchuster, Radhakrishnan Mahadevan, Jürgen Zanghellini, and Stefan Müller. Fromelementary flux modes to elementary flux vectors: Metabolic pathway analysis witharbitrary linear flux constraints. PLoS Comput.Biol., 13(4):e1005409, 2017.

Dhinakar S. Kompala and Ozturk S. Sadettin. Optimization of High Cell Density PerfusionBioreactors. In Cell Culture Technology for Pharmaceutical and Cell-Based Therapies,chapter 11, pages 387–416. CRC Press, Taylor & Francis Group, LLC, Boca Raton, FL,first edition, 2006.

Cleo Kontoravdi, Steven P. Asprey, Efstratios N. Pistikopoulos, and Athanasios Mantalaris.Development of a dynamic model of monoclonal antibody production and glycosylationfor product quality monitoring. Comput. Chem. Eng., 31:392–400, 2007.

Eric Langer. 2017 Biopharmaceutical Trends—Opportunities For TheNew Year, 2018. URL https://www.bioprocessonline.com/doc/

biopharmaceutical-trends-opportunities-for-the-new-year-0001.

Alvaro R. Lara, Laura A. Palomares, and Octavio T. Ramírez. Scale-down: Simulat-ing large-scale cultures in the laboratory. In Christoph Wittmann and James C. Liao,editors, Industrial Biotechnology: Products and Processes, chapter 2, pages 55–79.WileyâARVCH Verlag GmbH & Co. KGaA, first edition, 2017.

Olive Leavy. Therapeutic antibodies: Past, present and future. Nat. Rev. Immunol., 10:297,2010.

Gaspard Lequeux, Joeri Beauprez, Jo Maertens, Ellen Van Horen, Wim Soetaert, ErickVandamme, and Peter A. Vanrolleghem. Dynamic Metabolic Flux Analysis Demonstratedon Cultures Where the Limiting Substrate Is Changed from Carbon to Nitrogen and ViceVersa. J. Biomed. Biotechnol., 2010:ID: 621645, 19 pages, 2010.

Richard Levins. The Strategy of Model Building in Population Biology. Am. Sci., 54(4):421–431, 1966.

Amanda M. Lewis, Nicholas R. Abu-Absi, Michael C. Borys, and Zheng Jian Li. The useof ’Omics technology to rationally improve industrial mammalian cell line performance.Biotechnol. Bioeng., 113(1):26–38, 2016.

135

Page 154: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Nathan E. Lewis, Xin Liu, Yuxiang Li, Harish Nagarajan, George Yerganian, EdwardO’Brien, Aarash Bordbar, Anne M. Roth, Je↵rey Rosenbloom, Chao Bian, Min Xie,Wenbin Chen, Ning Li, Deniz Baycin-Hizal, Haythem Latif, Jochen Forster, Michael J.Betenbaugh, Iman Famili, Xun Xu, Jun Wang, and Bernhard Ø. Palsson. Genomiclandscapes of Chinese hamster ovary cell lines as revealed by the Cricetulus griseus draftgenome. Nat. Biotechnol., 31(8):759–765, 2013.

Feng Li, Natarajan Vijayasankaran, Amy (Yijuan) Shen, Robert Kiss, and Ashraf Amanullah.Cell culture processes for monoclonal antibody production. mAbs, 2(5):466–477, 2010.

Wolfram Liebermeister and Edda Klipp. Bringing metabolic networks to life: conveniencerate law and thermodynamic constraints. Theor. Biol. Med. Modell., 3:41, 2006a.

Wolfram Liebermeister and Edda Klipp. Bringing metabolic networks to life: integration ofkinetic, metabolic, and proteomic data. Theor. Biol. Med. Modell., 3:42, 2006b.

Francisco Llaneras and Jesús Picó. Stoichiometric Modelling of Cell Metabolism. J. Biosci.Bioeng., 105(1):1–11, 2008.

Francisco Llaneras and Jesús Picó. Which metabolic pathways generate and characterizethe flux space? A comparison among elementary modes, extreme pathways and minimalgenerators. J. Biomed. Biotechnol., 2010:ID: 753904, 13 pages, 2010.

Helder Lopes and Isabel Rocha. Genome-scale modeling of yeast: chronology, applicationsand critical perspectives. FEMS Yeast Res., 17(5):fox050, 2017.

Marco E. Lübbecke and Jacques Desrosiers. Selected Topics in Column Generation. Oper.Res., 53(6):1007–1023, 2005.

R. Mahadevan and C. H. Schilling. The e↵ects of alternate optimal solutions in constraint-based genome-scale metabolic models. Metab. Eng., 5:264–276, 2003.

D. E. Martens. Metabolic Flux Analysis of mammalian cells. In M. Al-Rubeai andM. Fussenegger, editors, Systems Biology, chapter 9, pages 275–299. Springer, 2007.

Verónica S. Martínez, Stefanie Dietmair, Lake-Ee Quek, Mark P. Hodson, Peter Gray, andLars K. Nielsen. Flux balance analysis of CHO cells before and after a metabolic switchfrom lactate production to consumption. Biotechnol. Bioeng., 110(2):660–666, 2013.

V. McMurray-Beaulieu, S. Hisiger, C. Durand, M. Perrier, and M. Jolicoeur. Na-butyratesustains energetic states of metabolism and t-PA productivity of CHO cells. J. Biosci.Bioeng., 108(2):160–167, 2009.

Leonor Michaelis and Maud Menten. Die Kinetik der Invertin-Wirkung (the kinetics ofinvertase activity). Biochem. Z., 49:333–369, 1913.

136

Page 155: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Jacques Monod. The Growth of Bacterial Cultures. Annu. Rev. of Microbiol., 3:371–394,1949.

Evelien Moorkens, Nicolas Meuwissen, Isabelle Huys, Paul Declerck, Arnold G. Vulto, andSteven Simoens. The Market of Biopharmaceutical Medicines: A Snapshot of a DiverseIndustrial Landscape. Front. Pharmacol., 8:314, 2017.

Rafael Moreno-Sánches, Emma Saavedra, Sara Rodríguez-Enríquez, and Viridiana Olín-Sandova. Metabolic Control Analysis : A Tool for Designing Strategies to. J. Biomed.Biotechnol., 2008:ID: 597913, 30 pages, 2008.

Bhanu Chandra Mulukutla, Salmaan Khan, Alex Lange, and Wei-Shou Hu. Glucosemetabolism in mammalian cell culture: new insights for tweaking vintage pathways.Trends Biotechnol., 28:476–484, 2010.

Bhanu Chandra Mulukutla, Michael Gramer, and Wei-Shou Hu. On metabolic shift tolactate consumption in fed-batch culture of mammalian cells. Metab. Eng., 14:138–49,2012.

Saeideh Naderi, Mukesh Meshram, Catherine Wei, Brendan McConkey, Brian Ingalls,Hector Budman, and Jeno Scharer. Development of a mathematical model for evaluatingthe dynamics of normal and apoptotic Chinese hamster ovary cells. Biotechnol. Prog., 27(5):1197–205, 2011.

D. Nelson and M. Cox. Lehninger Principles of Biochemistry. W. H. Freeman, New York,fourth edition, 2005.

Averina Nicolae, Judith Wahrheit, Janina Bahnemann, An-Ping Zeng, and Elmar Heinzle.Non-stationary (13)C metabolic flux analysis of Chinese hamster ovary cells in batch cul-ture using extracellular labeling highlights metabolic reversibility and compartmentation.BMC Syst. Biol., 8:50, 2014.

Jens Niklas and Elmar Heinzle. Metabolic Flux Analysis in Systems Biology of MammalianCells. In Genomics and Systems Biology of Mammalian Cell Culture, pages 109–132.Springer, Berlin, Heidelberg, 2011.

Jens Niklas, Konstantin Schneider, and Elmar Heinzle. Metabolic flux analysis in eukaryotes.Curr. Opin. Biotechnol., 21:63–69, 2010.

Jens Niklas, Eva Schräder, Volker Sandig, Thomas Noll, and Elmar Heinzle. Quantitativecharacterization of metabolism and metabolic shifts during growth of the new human cellline AGE1.HN using time resolved metabolic flux analysis. Bioprocess Biosyst. Eng., 34:533–45, 2011.

Yasu Nishiuch, Mieko Sasaki, Michie Nakayasu, and Atsushi Okiawa. Cytotoxicity ofcysteine in culture media. In Vitro, 12(9):635–638, 1976.

137

Page 156: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Hongxing Niu, Zakaria Amribt, Patrick Fickers, Wensong Tan, and Philippe Bogaerts. Meta-bolic pathway analysis and reduction for mammalian cell cultures—Towards macroscopicmodeling. Chem. Eng. Sci., 102:461–473, 2013.

Ryan P. Nolan and Kyongbum Lee. Dynamic model of CHO cell metabolism. Metab. Eng.,13:108–24, 2011.

Gregg B. Nyberg, R. Robert Balcarcel, Brian D. Follstad, Gregory Stephanopoulos, andDaniel I.C. Wang. Metabolism of peptide amino acids by Chinese hamster ovary cellsgrown in a complex medium. Biotechnol. Bioeng., 62:324–335, 1999.

Hildur Æsa Oddsdóttir. Macroscopic Modeling of Metabolic Reaction Networks andDynamic Identification of Elementary Flux Modes by Column Generation. Doctoralthesis, KTH Royal Institute of Technology, 2015.

Mehmet A. Orman, Francois Berthiaume, Ioannis P. Androulakis, and Marianthi G. Ier-apetritou. Advanced Stoichiometric Analysis of Metabolic Networks of MammalianSystems. Crit. Rev. Biomed. Eng., 39(6):511–534, 2011.

Je↵rey D Orth, Ines Thiele, and Bernhard Ø. Palsson. What is flux balance analysis? Nat.Biotechnol., 28(3):245–248, 2010.

Je↵rey D. Orth, Tom M. Conrad, Jessica Na, Joshua A. Lerman, Hojung Nam, Adam M.Feist, and Bernhard Ø. Palsson. A comprehensive genome-scale reconstruction ofEscherichia coli metabolism — 2011. Mol. Syst. Biol., 7:535, 2011.

Csaba Pál, Balázs Papp, and Martin J. Lercher. Adaptive evolution of bacterial metabolicnetworks by horizontal gene transfer. Nat. Genet., 37(12):1372–1375, 2005.

Jason A. Papin, Nathan D. Price, and Bernhard Ø. Palsson. Extreme Pathway Lengthsand Reaction Participation in Genome-Scale Metabolic Networks. Genome Res., 12:1889–1900, 2002.

Jason A Papin, Joerg Stelling, Nathan D Price, Ste↵en Klamt, Stefan Schuster, and Bern-hard Ø Palsson. Comparison of network-based pathway analysis methods. TrendsBiotechnol., 22(8):400–405, 2004.

Hyo-Soon Park, Ik-Hwan Kim, Ick-Young Kim, Ki-Ho Kim, and Hong-Jin Kim. Expressionof carbamoyl phosphate synthetase I and ornithine transcarbamoylase genes in Chinesehamster ovary dhfr-cells decreases accumulation of ammonium ion in culture media. J.Biotechnol., 81:129–140, 2000.

Rui Pereira, Jens Nielsen, and Isabel Rocha. Improving the flux distributions simulated withgenome-scale metabolic models of Saccharomyces cerevisiae. Metabol. Eng. Comm., 3:153–163, 2016.

138

Page 157: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Timothy M. Pierpont, Candice B. Limper, and Kristy L. Richards. Past, Present, and Futureof Rituximab—The World’s First Oncology Monoclonal Antibody Therapy. Front.Oncol., 8:163, 2018.

Ralf Pörtner, Uwe Jandt, and An-Ping Zeng. Cell Culture Technology. In ChristophWittmann and James C. Liao., editors, Industrial Biotechnology: Products and Processes,chapter 4, pages 129–158. Wiley-VCH Verlag GmbH & Co. KGaA, first edition, 2017.

A. Provost and G. Bastin. Dynamic metabolic modelling under the balanced growthcondition. J. Process Contr., 14:717–728, 2004.

A. Provost, G. Bastin, S.N. Agathos, and Y.-J. Schneider. Metabolic Design of MacroscopicModels: Application to CHO cells. In Proceedings of the 44th IEEE Conference onDecision and Control, and the European Control Conference 2005, pages 2982–2989,Seville, Spain, 2005.

Agnés Provost. Metabolic Design of Dynamic Bioreaction Models. Doctoral thesis, Univer-sité Catholique de Louvain, 2006.

T. Puck, Theodore, J. Cieciura, Steven, and Arthur Robinson. Genetics of somatic mam-malian cells III. Long-term Cultivation of Euploid cells from Human and Animal Subjects.J. Exp. Med., 108:945–956, 1958.

Lake-Ee Quek, Stefanie Dietmair, Jens O. Krömer, and Lars K. Nielsen. Metabolic fluxanalysis in mammalian cell culture. Metab. Eng., 12:161–171, 2010.

R. A. Rader and E. S. Langer. Thirty Years of Upstream Productivity Im-provements. BioProcess International, pages 10–12, 2015. URL https:

//bioprocessintl.com/upstream-processing/expression-platforms/

30-years-upstream-productivity-improvements/.

Živa Rejc, Lidija Magdevska, Tilen Tršelic, Timotej Osolin, Rok Vodopivec, Jakob Mraz,Eva Pavliha, Nikolaj Zimic, Tanja Cvitanovic, Damjana Rozman, Miha Moškon, andMiha Mraz. Computational modelling of genome-scale metabolic networks and itsapplication to CHO cell cultures. Comput. Biol. Med., 88:150–160, 2017.

Julien Robitaille, Jingkui Chen, and Mario Jolicoeur. A Single Dynamic Metabolic ModelCan Describe mAb Producing CHO Cell Batch and Fed-Batch Cultures on Di↵erentCulture Media. PLoS ONE, 10(9):e0136815, 2015.

Hendrik Rohn, Anja Hartmann, Astrid Junker, Björn H. Junker, and Falk Schreiber.FluxMap: A VANTED add-on for the visual exploration of flux distributions in bi-ological networks. BMC Syst. Biol., 6:33, 2012.

139

Page 158: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Thomas P. Ryan. Design of experiments. In David J. Balding, Noel A. C. Cressie, Garrett M.Fitzmaurice, Harvey Goldstein, Iain M. Johnstone, Geert Molenberghs, David W. Scott,Adrian F. M. Smith, Ruey S. Tsay, Sanford Weisberg, Vic Barnett, J. Stuart Hunter,Joseph B. Kadane, and Jozef L. Teugels, editors, Wiley Series in Probability and Statistics;840: Statistical Methods for Quality Improvement, chapter 13, pages 435–511. JohnWiley & Sons, Inc., Hoboken, New Jersey, third edition, 2011.

C. S. Sanderson, P. J. Phillips, and J. P. Barford. Structured modelling of animal cells.Cytotechnology, 21:149–153, 1996.

Filipe Santos, Joost Boele, and Bas Teusink. A Practical Guide to Genome-Scale MetabolicModels and Their Analysis. In Daniel Jameson, Malkhey Verma, and Hans V. Westerho↵,editors, Methods Enzymol. Vol. 500, volume 500, chapter 24, pages 509–532. Burlington:Academic Press, 2011.

Jan Schellenberger, Richard Que, Ronan M. T. Fleming, Ines Thiele, Je↵rey D. Orth,Adam M. Feist, Daniel C. Zielinski, Aarash Bordbar, Nathan E. Lewis, Sorena Rahma-nian, Joseph Kang, Daniel R. Hyduke, and Bernhard Ø. Palsson. Quantitative predictionof cellular metabolism with constraint-based models: the COBRA Toolbox v2.0. Nat.Protocols, 6(9):1290–1307, 2011.

David W. Schryer, Pearu Peterson, Toomas Paalme, and Marko Vendelin. Bidirectionalityand compartmentation of metabolic fluxes are revealed in the dynamics of isotopomernetworks. Int. J. Mol. Sci., 10:1697–1718, 2009.

Robert Schuetz, Lars Kuepfer, and Uwe Sauer. Systematic evaluation of objective functionsfor predicting intracellular fluxes in Escherichia coli. Mol. Syst. Biol., 3:119, 2007.

Stefan Schuster and Claus Hilgetag. On Elementary Flux Modes in Biochemical ReactionSystems at Steady State. J. Biol. Syst., 2(2):165–182, 1994.

T. Seewöster and J. Lehmann. Influence of targeted asparagine starvation on extra- andintracellular amino acid pools of cultivated Chinese hamster ovary cells. Appl. Microbiol.Biotechnol., 44:344–350, 1995.

Christopher A. Sellick, Alexandra S. Croxford, Arfa R. Maqsood, Gill Stephens, Hans V.Westerho↵, Royston Goodacre, and Alan J. Dickson. Metabolite profiling of recombinantCHO cells: designing tailored feeding regimes that enhance recombinant antibodyproduction. Biotechnol. Bioeng., 108(12):3025–3031, 2011.

Suresh Selvarasu, Iftekhar A. Karimi, Ghi-Hoon Ghim, and Dong-Yup Lee. Genome-scalemodeling and in silico analysis of mouse cell metabolic network. Mol. BioSyst., 6:152–161, 2010.

140

Page 159: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Suresh Selvarasu, Ying Swan Ho, William P. K. Chong, Niki S. C. Wong, Faraaz N. K.Yusufi, Yih Yean Lee, Miranda G. S. Yap, and Dong-Yup Lee. Combined in silico model-ing and metabolomics analysis to characterize fed-batch CHO cell culture. Biotechnol.Bioeng., 109(6):1415–1429, 2012.

Neelanjan Sengupta, Steven T. Rose, and John A. Morgan. Metabolic flux analysis of CHOcell metabolism in the late non-growth phase. Biotechnol. Bioeng., 108(1):82–92, 2011.

Kashif Sheikh, Jochen Förster, and Lars K. Nielsen. Modeling hybridoma cell metabolismusing a generic genome-scale metabolic model of Mus musculus. Biotechnol. Prog., 21(1):112–21, 2005.

Yury Sidorenko, Aljoscha Wahl, Michael Dauner, Yvonne Genzel, and Udo Reichl. Com-parison of metabolic flux distributions for MDCK cell growth in glutamine- and pyruvate-containing media. Biotechnol. Prog., 24:311–320, 2008.

Sigma-Aldrich Media Expert. Cysteine in Cell Culture. URL https:

//www.sigmaaldrich.com/life-science/cell-culture/learning-center/

media-expert/cysteine.html.

Martin I. Sigurdsson, Neema Jamshidi, Eirikur Steingrimsson, Ines Thiele, and Bernhard Ø.Palsson. A detailed genome-wide reconstruction of mouse metabolism based on humanRecon 1. BMC Syst. Biol., 4:140, 2010.

Peter Soelkner. Past, Present, and Future Trends A↵ecting the Bio/Pharmaceutical Industry,2017. URL http://www.pharmtech.com/print/329185?page=full.

Si Nga Sou, Christopher Sellick, Ken Lee, Alison Mason, Sarantos Kyriakopoulos, Karen M.Polizzi, and Cleo Kontoravdi. How does mild hypothermia a↵ect monoclonal antibodyglycosylation? Biotechnol. Bioeng., 112(6):1165–1176, 2015.

Ralf Steuer and Björn H. Junker. Computational Models of Metabolism: Stability andRegulation in Metabolic Networks. Adv. Chem. Phys., 142, 2009.

Jure Strnad, Matjaž Brinc, Vatroslav Spudic, Nadja Jelnikar, and Lidija Mirnik. Optimizationof Cultivation Conditions in Spin Tubes for CHO Cells Producing Erythropoietin. InS. Pierucci and G. Buzzi Ferraris, editors, 20th European Symposium on Computer AidedProcess Engineering — ESCAPE20. Elsevier B.V., 2010.

Tamás Székely and Kevin Burrage. Stochastic simulation in systems biology. Comput.Struct. Biotechnol. J., 12:14–25, 2014.

Marco Terzer and Jörg Stelling. Large-scale computation of elementary flux modes with bitpattern trees. Bioinformatics, 24(19):2229–2235, 2008.

141

Page 160: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Kathryn E. Tiller and Peter M. Tessier. Advances in Antibody Design. Annu. Rev. Biomed.Eng., 17:191–216, 2015.

H. M. Tsuchiya, A. G. Fredrickson, and R. Aris. Dynamics of Microbial Cell Populations.Adv. Chem. Eng., 6:125–206, 1966.

R. Urbanczik and C. Wagner. An improved algorithm for stoichiometric network analysis:theory and applications. Bioinformatics, 21(7):1203–1210, 2005.

Sabine Vclear, Melcher Michael, Norbert Auer, Astrid Hrdina, Anja Puklowski, FriedrichLeisch, Vaibhav Jadhav, Till Wenger, Martina Baumann, and Nicole Borth. Changes inChromosome Counts and Patterns in CHO Cell Lines upon Generation of RecombinantCell Lines and Subcloning. Biotechnol. J., 13:1700495, 2018.

M. Vivian, Lisa R. Abston, Albert E. Schmelzer, Shinie Shaw, and William M. Miller.Selected Amino Acids Protect Hybridoma and CHO Cells from Elevated Carbon Dioxideand Osmolality. Biotechnol. Bioeng., 78(7):741–752, 2002.

Axel von Kamp and Stefan Schuster. Metatool 5.0: fast and flexible elementary modesanalysis. Bioinformatics (Oxford, England), 22(15):1930–1931, 2006.

Hendrik Waegeman and Wim Soetaert. Increasing recombinant protein production in Es-cherichia coli through metabolic and genetic engineering. J. Ind. Microbiol. Biotechnol.,38:1891–1910, 2011.

Judith Wahrheit, Averina Nicolae, and Elmar Heinzle. Eukaryotic metabolism: measuringcompartment fluxes. Biotechnol. J., 6:1071–1085, 2011.

Judith Wahrheit, Averina Nicolae, and Elmar Heinzle. Dynamics of growth and metabolismcontrolled by glutamine availability in Chinese hamster ovary cells. Appl. Microbiol.Biotechnol., 98:1771–1783, 2014a.

Judith Wahrheit, Jens Niklas, and Elmar Heinzle. Metabolic control at the cytosol–mitochondria interface in di↵erent growth phases of CHO cells. Metab. Eng., 23:9–21,2014b.

W. D. Wallis. Linear Programming. In A Beginner’s Guide to Finite Mathematics: ForBusiness, Management, and the Social Sciences, chapter 6, pages 281–357. BirkhäuserBoston, second edition, 2012.

Gary Walsh. Biopharmaceutical benchmarks 2014. Nat. Biotechnol., 32(10):992–1002,2014.

Otto Warburg. On the origin of cancer cells. Science, 123(3191):309–314, 1956.

Michael Weisberg. Three Kinds of Idealization. J. Philos., 104(12):639–659, 2007.

142

Page 161: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Christoph Wittmann, James C. Liao, Hans-Peter Meyer, Wolfgang Minas, and DiegoSchmidhalter. Industrial-Scale Fermentation. In Christoph Wittmann and James C. Liao,editors, Industrial Biotechnology: Products and Processes, pages 1–53. Wiley-VCHVerlag GmbH & Co. KGaA., first edition, 2017.

Florian M. Wurm. CHO Quasispecies—Implications for Manufacturing Processes. Pro-cesses, 1:296–311, 2013.

Florian M. Wurm and David Hacker. First CHO genome. Nat. Biotechnol., 29(8):718–720,2011.

Florian M. Wurm, David L. Hacker, and Sophie Nallet. Recombinant protein productionyields from mammalian cells: past, present, and future. Guide to Protein Production.BioPharm Int., 2008(5), 2008.

Liangzhi Xie and Daniel I. C. Wang. Fed-Batch Cultivation of Animal Cells Using Di↵erentMedium Design Concepts and Feeding Strategies. Biotechnol. Bioeng., 95(2):270–284,1994a.

Liangzhi Xie and Daniel I. C. Wang. Material balance studies on animal cell metabolismusing a stoichiometrically based reaction network. Biotechnol. Bioeng., 52:579–590,1996a.

Liangzhi Xie and Daniel I. C. Wang. Energy metabolism and ATP balance in animal cellcultivation using a stoichiometrically based reaction network. Biotechnol. Bioeng., 52:591–601, 1996b.

Liangzhi Xie and Daniel I.C. Wang. Stoichiometric analysis of animal cell growth and itsapplication in medium design. Biotechnol. Bioeng., 43:1164–1174, 1994b.

Liangzhi Xie and Weichang Zhou. Fed-Batch Cultivation of Mammalian Cells for theProduction of Recombinant Proteins. In Sadettin S. Ozturk and Wei-Shou Hu, editors,Cell Culture Technology for Pharmaceutical and Cell-Based Therapies, chapter 10, pages349–386. Taylor & Francis Group, LLC, Boca Raton, FL, first edition, 2006.

Zizhuo Xing, Brian Kenty, Inna Koyrakh, Michael Borys, Shih-Hsie Pan, and Zheng JianLi. Optimizing amino acid composition of CHO cell culture media for a fusion proteinproduction. Process Biochem., 46:1423–1429, 2011.

Ningning Xu, Chao Ma, Jianfa Ou, Wanqi Wendy Sun, Lufang Zhou, Hui Hu, and Xi-aoguang Margaret Liu. Comparative proteomic analysis of three Chinese hamster ovary(CHO) host cells. Biochem. Eng. J., 124:122–129, 2017.

Xun Xu, Harish Nagarajan, Nathan E. Lewis, Shengkai Pan, Zhiming Cai, Xin Liu, WenbinChen, Min Xie, Wenliang Wang, Stephanie Hammond, Mikael R. Andersen, Norma

143

Page 162: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

References

Ne↵, Benedetto Passarelli, Winston Koh, H. Christina Fan, Jianbin Wang, Yaoting Gui,Kelvin H. Lee, Michael J. Betenbaugh, Stephen R. Quake, Iman Famili, Bernhard Ø.Palsson, and Jun Wang. The genomic sequence of the Chinese hamster ovary (CHO)-K1cell line. Nat. Biotechnol., 29(8):735–741, 2011.

F. Zamorano, A. Vande Wouwer, and G. Bastin. A detailed metabolic flux analysis of anunderdetermined network of CHO cells. J. Biotechnol., 150:497–508, 2010.

F. Zamorano, A. Vande Wouwer, R. M. Jungers, and G. Bastin. Dynamic metabolic modelsof CHO cell cultures through minimal sets of elementary flux modes. J. Biotechnol., 164:409–422, 2013.

Francisca Zamorano Riveros. Metabolic Flux Analysis of CHO cell cultures. Doctoralthesis, University of Mons, 2012.

Jürgen Zanghellini, David E. Ruckerbauer, Michael Hanscho, and Christian Jungreuthmayer.Elementary flux modes in a nutshell: Properties, calculation and applications. Biotechnol.J., 8:1009–1016, 2013.

An-Ping Zeng and Jing-xiu Bi. Cell Culture Kinetics and Modeling. In Sadettin S. Ozturkand Wei-Shou Hu, editors, Cell Culture Technology for Pharmaceutical and Cell-BasedTherapies, chapter 9, pages 299–348. CRC Press, Taylor & Francis Group, LLC, BocaRaton, FL, first edition, 2006.

Jinyou Zhang. Mammalian Cell Culture for Biopharmaceutical Production. In Richard H.Baltz, Julian E. Davies, and Arnold L. Demain, editors, Manual of Industrial Microbiol-ogy and Biotechnology, chapter 12, pages 157–178. ASM Press, Washington, DC., thirdedition, 2010.

Ye Zhang, Per Stobbe, Christian Orrego Silvander, and Véronique Chotteau. Very highcell density perfusion of CHO cells anchored in a non-woven matrix-based bioreactor. J.Biotechnol., 213:28–41, 2015.

Meixia Zhou, Yongping Crawford, Domingos Ng, Jack Tung, Abigail F.J. Pynn, AngelaMeier, Inn H. Yuk, Natarajan Vijayasankaran, Kimberly Leach, John Joly, BradleySnedecor, and Amy Shen. Decreasing lactate level and increasing antibody production inChinese Hamster Ovary cells (CHO) by reducing the expression of lactate dehydrogenaseand pyruvate dehydrogenase kinases. J. Biotechnol., 153:27–34, 2011.

Craig Zupke and Gregory Stephanopoulos. Intracellular flux analysis in hybridoma usingmass balances and in vitro 13C NMR. Biotechnol. Bioeng., 45:292–303, 1995.

144

Page 163: Macroscopic models of Chinese hamster ovary cell cultureskth.diva-portal.org/smash/get/diva2:1362872/FULLTEXT01.pdf · Mathematical modeling has great potential to speed up this work

Acknowledgments

The realization of this thesis was made possible thanks to the Excellence ScholarshipFunding for doctoral studies provided by KTH and the School of Biotechnology,and research grants from Vinnova.

I thank my supervisor Dr. Véronique Chotteau, for the support and guidanceprovided throughout the entire doctoral education. I would also like to thankour collaborators Dr. Hildur Æsa Oddsdóttir and Prof. Anders Forsgren from theDepartment of Mathematics at KTH. Hildur Æsa has since she received her PhDin Applied and Computational Mathematics supported my continued work and thefinalization of this thesis. Anders Forsgren, the main supervisor of Hildur Æsa, hasco-supervised my project, and continuously supported our collaboration.

I would like to thank Irvine Scientific (USA) who provided the chemically-defined cultivation medium, and Selexis (Switzerland) who kindly provided theindustrial CHO cell line. These two contributions from the industry were veryimportant for bringing my experimental work forward.

I would like to thank the colleagues at the Department of Industrial Biotechnologyand in the Cell Technology Group that I have met and come to know over the years.I especially thank Atefeh Shokri who is responsible for the cell culture laboratory ofthe Cell Culture Technology Group at KTH at which I carried out the experimentalwork, Jan Kinnander who helped me with setting up and carrying out the aminoacid analyses, and Ye Zhang who helped me to get started with the IgG analyses.

I thank Eva and Anne for providing me quiet places to sit and write, and Evafor her help with the final editing of the text.

Finally, I thank my family: Joakim, Leonard, and Waldemar. For alwayssupporting me and reminding me of who I am. And for waiting so patiently for meto complete this "book".

145