27.02.2013 Views

Download PDF - BioMed Central

Download PDF - BioMed Central

Download PDF - BioMed Central

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

MEETING ABSTRACTS Open Access<br />

22nd European Society for Animal Cell Technology<br />

(ESACT) Meeting on Cell Based Technologies<br />

Vienna, Austria. 15-18 May 2011<br />

Edited by Hansjörg Hauser<br />

Published: 22 November 2011<br />

These abstracts are available online at http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

INTRODUCTION<br />

I1<br />

Cell Based Technologies: Abstracts of the 22 nd ESACT Meeting 2011<br />

in Vienna<br />

Hansjörg Hauser<br />

Helmholtz-Zentrum für Infektionsforschung GmbH, Department of Gene<br />

Regulation and Differentiation, 38124 Braunschweig, Germany<br />

E-mail: hansjoerg.hauser@helmholtz-hzi.de<br />

BMC Proceedings 2011, 5(Suppl 8):I1<br />

The European Society of Animal Cell Technology (ESACT) is a society that<br />

brings together scientists, engineers and other specialists working with<br />

animal cells in order to promote communication of experiences between<br />

European and international investigators and progress development of cell<br />

systems in productions derived from them.<br />

Animal cells are being used as substrates in basic research and also for the<br />

production of proteins. Tissue engineering, gene and cell therapies, organ<br />

replacement technologies and cell-based biosensors contribute to a<br />

considerable widening of interest and research activity based on animal cell<br />

technology.<br />

The abstracts of this supplement are from the 22 nd ESACT meeting that was<br />

held in Vienna, Austria, May 15-18, 2011. The abstracts review the<br />

presentations from this meeting and should be a useful resource for the<br />

state-of-the-art in animal cell technology.<br />

ORAL PRESENTATIONS<br />

O1<br />

Highly efficient, chemically defined and fully scalable biphasic<br />

production of vaccine viruses<br />

Ingo Jordan * , Volker Sandig<br />

ProBioGen AG, 13086 Berlin, Germany<br />

E-mail: ingo.jordan@probiogen.de<br />

BMC Proceedings 2011, 5(Suppl 8):O1<br />

Vectorial vaccines are predicted to yield novel therapeutic and protective<br />

approaches. They consist of recombinant live carriers that express antigen<br />

from an unrelated pathogen in the recipient. Promising viral carriers are hostrestricted<br />

pox viruses that trigger a strong immune response without ability<br />

to replicate in the human organism. A block in replication is an important<br />

safety feature that allows application even in immunocompromized<br />

recipients. However, with this type of attenuation there is not even limited<br />

amplification of the vector at the site of infection and therefore very high<br />

numbers of infectious units have to be given per dose for full efficacy. Hence,<br />

if such vectors are to be used in global vaccine programs highly efficient<br />

production processes will be required. Furthermore, to combat diseases such<br />

as AIDS, hepatitis C, tuberculosis, or malaria with these vectors, millions of<br />

the concentrated vaccine units will have to be provided annually. Any<br />

production process therefore should also be scalable and preferrably<br />

transferrable to newly industrialized countries. Latter requirement demands a<br />

robust process independent of the complex logistics and uncertainties<br />

associated with primary chicken cells, the current industrial substrate for<br />

these pox viruses and certain other vectors.<br />

Webelievethatwehavesolvedmostoftheupstreamchallengeswitha<br />

chemically defined suspension culture production process for three<br />

disparate members of the highly attenuated poxviruses [1]: modified<br />

vaccinia Ankara (MVA), fowlpoxvirus (FPV) and canarypoxvirus ALVAC. The<br />

process is independent of primary material and based on the continuous<br />

duck suspension cell line AGE1.CR specifically created as a vaccine substrate<br />

[2].<br />

In contrast to production of influenza virus that readily replicates in the cell<br />

proliferation medium [3], development of a production process for<br />

poxviruses was surprisingly complicated and involves media formulations<br />

matched to two distinct phases, cell proliferation and virus production. Our<br />

process was adjusted to the different kinetics and requirements of the<br />

three examined viruses, and was studied in Wave and disposable<br />

bioreactors up to 50 L scale. For MVA, titers in the crude lysate without any<br />

processing reliably exceed the critical threshold of 10 8 pfu/mL and often<br />

are in the range of 5 × 10 8 to 2 × 10 9 pfu/mL.<br />

One hallmark of the biphasic process described here is controlled formation<br />

of suspension cell aggregates at the transition from cell proliferation to virus<br />

production. Such aggregate formation was achieved with distinct chemically<br />

defined media harmonized such that a highly efficient, robust and industrial<br />

biphasic processes was obtained that does not require perfusion, medium<br />

replacement or microcarriers.<br />

In addition to poxviruses, this approach was successful in production of an<br />

unrelated RNA vector and also for this reason we believe that we have<br />

developed a more general principle for scalable production of viruses that<br />

may benefit from cell-to-cell contacts: In the background of the AGE1.CR cell<br />

lines, packaging cells were created for SIN/VEE-chimeric alphavirus replicons<br />

[4]. The packaging cells were transfected for stable trans-complementation<br />

of envelope and capsid proteins from separate expression cassettes.<br />

Production of replicons was efficient using adherent (serum-dependent)<br />

cultures but only moderately efficient with suspension cultures of the<br />

packaging cells. After transfer and optimization of the pox virus production<br />

process to replicon-induced packaging cell cultures (shown in figure 1) we<br />

obtained yields beyond 10 8 pfu/mL.<br />

Appearance of suspension packaging cells just prior to infection and at<br />

various time points during virus production is shown in (A). Note induction of<br />

aggregates in presence of virus production medium and cytopathic effect<br />

24 h post induction. Yields of replicon is shown in (B) with peak titers 24 h to<br />

48 h post induction.<br />

© 2011 various authors, licensee <strong>BioMed</strong> <strong>Central</strong> Ltd. All articles published in this supplement are distributed under the terms of the<br />

Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and<br />

reproduction in any medium, provided the original work is properly cited.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract O1) Biphasic process adapted to alphavirus replicons.<br />

In summary, superior yields were obtained for unrelated viral vectors<br />

using separate chemically defined media for proliferation and virus<br />

production. The media are matched to allow highly efficient, robust and<br />

industrial biphasic processes that do not require perfusion or medium<br />

replacement.<br />

References<br />

1. Jordan I, Northoff S, Thiele M, Hartmann S, Horn D, Höwing K, Bernhardt H,<br />

Oehmke S, von Horsten H, Rebeski D, Hinrichsen L, Zelnik V, Mueller W,<br />

Sandig V: A chemically defined production process for highly attenuated<br />

poxviruses. Biologicals 2011, 39:50-58.<br />

2. Jordan I, Vos A, Beilfuss S, Neubert A, Breul S, Sandig V: An avian cell line<br />

designed for production of highly attenuated viruses. Vaccine 2009,<br />

27:748-56.<br />

3. Lohr V, Rath A, Genzel Y, Jordan I, Sandig V, Reichl U: New avian<br />

suspension cell lines provide production of influenza virus and MVA in<br />

serum-free media: studies on growth, metabolism and virus<br />

propagation. Vaccine 2009, 27:4975-82.<br />

4. Perri S, Greer CE, Thudium K, Doe B, Legg H, Liu H, Romero RE, Tang Z,<br />

Bin Q, Dubensky TW Jr, Vajdy M, Otten GR, Polo JM: An alphavirus replicon<br />

particle chimera derived from venezuelan equine encephalitis and<br />

sindbis viruses is a potent gene-based vaccine delivery vector. J Virol<br />

2003, 77:10394-403.<br />

O2<br />

Recombinant antibody mixtures; optimization of cell line generation<br />

and single-batch manufacturing processes<br />

Søren K Rasmussen * , Lars S Nielsen, Christian Müller, Thomas Bouquin,<br />

Henrik Næsted, Nina T Mønster, Frank Nygaard, Dietmar Weilguny,<br />

Torben P Frandsen, Anne B Tolstrup<br />

Symphogen A/S, Elektrovej 375, 2800 Lyngby, Denmark<br />

E-mail: skr@symphogen.com<br />

BMC Proceedings 2011, 5(Suppl 8):O2<br />

Background: Recombinant antibody mixtures represent an important new<br />

class of antibody therapeutics as demonstrated by the increasing amount<br />

of literature showing that combinations of two or more antibodies show<br />

superiority compared to monoclonal antibodies (mAbs) for treatment of<br />

cancer and infectious diseases [1-5]. Sym004, composed of two antibodies<br />

targeting non-overlapping epitopes of the epidermal growth factor<br />

receptor (EGFR) act in a synergistic manner to induce an efficient<br />

internalization of EGFR leading to subsequent degradation and exhibit<br />

superior anticancer efficacy as demonstrated in several preclinical in vivo<br />

models [5].<br />

At Symphogen A/S, we have developed an expression platform, Sympress,<br />

for cost-efficient production of antibody mixtures. The antibody mixtures are<br />

produced using a single-batch manufacturing approach where a polyclonal<br />

working cell bank (pWCB) prepared by mixing the individual stable cell lines<br />

producing all the desired antibodies is used as seed material for a bioreactor<br />

process [6]. By using a single-batch approach the CMC development costs of<br />

Page 2 of 181<br />

antibody mixtures are comparable to costs for monoclonal antibodies.<br />

However, the single-batch manufacturing approach raises questions with<br />

regard to control of composition ratios, compositional stability and<br />

robustness of the cell banking procedure.<br />

Here, we present experimental data addressing these key questions and<br />

demonstrate that mixtures of recombinant antibodies can be produced<br />

under predictable, reproducible and stable conditions using the<br />

Sympress technology.<br />

Material and methods: The second generation Sympress technology is<br />

based on expression in the ECHO cell line, a genetically modified version<br />

of the dihydrofolate reductase (DHFR) negative Chinese Hamster Ovary<br />

(CHO) cell line DG44 [7].<br />

The expression plasmid used for stable transfection contained a<br />

bidirectional CMV promoter construct enabling co-expression of the IgG<br />

light and heavy chains from one plasmid. A DHFR selection marker was<br />

coupled directly to the heavy chain via an internal ribosome entry site<br />

(IRES).<br />

ECHO parental cells were transfected separately with each of the individual<br />

antibody expression vectors using standard transfection technology,<br />

whereafter cells were subjected to a methotrexate (MTX) selection schedule.<br />

The selected stable pools were single-cell cloned by FACS and highexpressing<br />

clones were adapted to chemically defined cell culture medium,<br />

expanded and frozen, still as individual monoclonal cell lines.<br />

The preparation of polyclonal cell banks followed a two-tiered cell<br />

banking approach. First, the relevant monoclonal cell lines were thawed<br />

and expanded. They were then mixed in predefined ratios and frozen as<br />

polyclonal master cell banks (pMCB). The pMCB was subsequently<br />

thawed, expanded and frozen as polyclonal working cell bank (pWCB).<br />

Antibody purification was performed by capture on a MabSelect SuRe<br />

column and cation exchange chromatography (CIEX) was used to<br />

separate the different antibodies based on their charge differences. The<br />

characteristics of the chromatograms (peak area/peak height) were used<br />

to determine the relative distribution of the different antibodies in the<br />

mixtures.<br />

Results: Two ECHO cell lines producing two distinct antibodies were<br />

selected based on their growth and production characteristics for a study to<br />

examine how mixing of the cells at different ratios affected the antibody<br />

composition. Briefly, the two cell lines were thawed, expanded and mixed in<br />

five different rations (5:5; 4:6; 3:7; 2:8 and 1:9). The resulting mixtures were<br />

frozen as pMCBs. To examine the obtained ratios and the compositional<br />

stability over prolonged periods of cultivation the pMCBs were revived and<br />

subjected to six weeks cultivation in shakers, followed by a 14 days fed<br />

batch process in shakers. Supernatant samples were harvested once a week<br />

and at the end of the fed batch process. IgG was captured by protein A and<br />

the relative antibody ratio was determined by CIEX. The results clearly<br />

showed that all the compositions were very stable over time and,<br />

importantly, that the relative amount of each antibody could be controlled<br />

by mixing the cell lines in an appropriate ratio (Figure 1A). Furthermore, the<br />

was a very strong correlation (R2 = 0.997) between expected and measured<br />

percentage of Ab 1 as shown in figure 1B.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract O2) A: Relative antibody ratios in pMCBs generated by mixing two ECHO cell lines at five different ratios. The antibody ratios were<br />

determined once a week by CIEX (from week 1 to 6 of cultivation) and in the harvest of a 14-day fed batch cultivation initiated after 6 weeks of<br />

cultivation. Red lines represent antibody 1 (Ab 1) and blue lines represent antibody 2 (Ab 2). B: Expected percentages plotted against measured<br />

percentages of Ab 1 in the five different mixes after 6 weeks seed train. C: Antibody compositions in pWCB in fed batch shaker experiments after 14, 21<br />

and 28 days seed train, respectively. D:Antibody distributions in harvest from seven different 5 L bioreactor processes. Four pWCB-1 ampoules (blue<br />

columns) and three pWCB-2 ampoules (green columns) were used for seed train. The lighter colored columns represent mean ± sdev.<br />

We then examined the compositional stability in a more complex model<br />

composed of a mixture of six different antibodies. A seed train of<br />

approximately 3 weeks would be required to generate cells for<br />

inoculation of production reactors in a 10.000 L scaled-up manufacturing<br />

Page 3 of 181<br />

process. To examine the robustness of the relative cell compositions<br />

during this timeframe +/- one week the cells from pWCB were inoculated<br />

into a 14 days fed batch shaker after 14, 21 or 28 days of expansion,<br />

respectively. The antibody distributions at the three different time points


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

were very constant as illustrated in Figure 1C. Additionally, the growth<br />

and production properties were also very constant over time (data not<br />

shown).<br />

The reproducibility and robustness of the cell banking system was<br />

evaluated by comparing the antibody product composition produced from<br />

two different pWCBs generated from the same pMCB. A pMCB producing a<br />

mixture containing 6 antibodies was used for the study. We generated two<br />

polyclonal working cell banks (pWCB-1 and pWCB-2) from the same pMCB.<br />

Production of pWCB-2 was separated in time from production of pWCB-1<br />

by several months. Four ampoules from pWCB-1 were expanded and used<br />

for inoculation of four 5 L bioreactors for fed batch production. Several<br />

months later, three ampoules from pWCB-2 were expanded and used to<br />

perform three new 5 L bioreactor runs. The antibodies were purified and<br />

antibody distribution determined by CIEX. The observed variation, both<br />

within the same pWCB and between different pWCB in regard to the<br />

antibody distributing was very limited (Figure 1D). This strongly indication<br />

that the pMCB/pWCB concept provides a reliable cell banking strategy. The<br />

reproducibility was also confirmed with respect to cell growth and<br />

productivity (data not shown).<br />

Conclusion: Symphogen A/S has developed an expression platform,<br />

Sympress, that can be used for predictable, reproducible, and stable<br />

production of antibody mixtures in a cost effective setting.<br />

Here, we have shown that the relative antibody ratio in antibody mixtures<br />

can be effective controlled using appropriate mixing of the individual<br />

monoclonal cell lines before generation of the pMCB. Further, we have<br />

presented consistent data from fed batch productions with pWCB where the<br />

seed train lengths varied from 14 to 28 days. This strongly supports that the<br />

single-batch manufacturing concept is sufficiently robust for manufacturing,<br />

also at scales required for commercial manufacturing. The two-tiered cell<br />

bank approach composed of pMCB and pWCB is robust and reproducible,<br />

and showed very high consistency both within a given pWCB and between<br />

different polyclonal working cell banks generated from the same pMCB.<br />

References<br />

1. Ben-Kasus T, Schechter B, Lavi S, Yarden Y, Sela M: Persistent elimination<br />

of ErbB-2/HER2-overexpressing tumors using combinations of<br />

monoclonal antibodies: Relevance of receptor endocytosis. Proc Natl<br />

Acad Sci U S A 2009, 106:3294-3299.<br />

2. van der Horst EH, Chinn L, Wang M, Velilla T, Tran H, Madrona Y, Lam A,<br />

Ji M, Hoey TC, Sato AK: Discovery of fully human anti-MET monoclonal<br />

antibodies with antitumor activity against colon cancer tumor models in<br />

vivo. Neoplasia 2009, 11:355-364.<br />

3. Dong J, Demarest SJ, Sereno A, Tamraz S, Langley E, Doern A, Snipas T,<br />

Perron K, Joseph I, Glaser SM, Ho SN, Reff ME, Hariharan K: Combination of<br />

two insulin-like growth factor-I receptor inhibitory antibodies targeting<br />

distinct epitopes leads to an enhanced antitumor response. Mol Cancer<br />

Ther 2010, 9:2593-2604.<br />

4. Fuentes G, Scaltriti M, Baselga J, Verma CS: Synergy between trastuzumab<br />

and pertuzumab for human epidermal growth factor 2 (Her2) from<br />

colocalization: an in silico based mechanism. Breast Cancer Res 2011 in<br />

press.<br />

5. Pedersen MW, Jacobsen HJ, Koefoed K, Hey A, Pyke C, Haurum JS, Kragh M:<br />

Sym004: Novel synergistic anti-epidermal growth factor receptor<br />

antibody mixture with superior anticancer efficacy. Cancer Res 2010,<br />

15:588-597.<br />

6. Frandsen TP, Naested H, Rasmussen SK, Hauptig P, Wiberg FC,<br />

Rasmussen LK, Jensen AM, Persson P, Wikén M, Engström A, Jiang Y,<br />

Thorpe SJ, Förberg C, Tolstrup AB: Consistent manufacturing and quality<br />

control of a highly complex recombinant polyclonal product for human<br />

therapeutic use. Biotech and Bioeng 2011 in press.<br />

7. Nielsen LS, Baer A, Müller C, Gregersen K, Mønster NT, Rasmussen SK,<br />

Weilguny D, Tolstrup AB: Single-batch production of recombinant human<br />

polyclonal antibodies. Mol Biotechnol 2010, 45:257-266.<br />

O3<br />

Cell-based medicinal products and the development of GMP-compliant<br />

processes and manufacturing<br />

Luca Romagnoli * , Ilaria Giuntini, Marta Galgano, Chiara Crosta,<br />

Luigi Cavenaghi, Maria Luisa Nolli<br />

Areta International s.r.l., Gerenzano (VA), 21040, Italy<br />

E-mail: lromagnoli@aretaint.com<br />

BMC Proceedings 2011, 5(Suppl 8):O3<br />

Page 4 of 181<br />

When performing the GMP process development and scale up of cellular<br />

therapies, a critical review of the manufacturing process and all the<br />

materials and reagents involved in the production steps is the mandatory<br />

starting point to avoid potential issues related to the quality and safety of<br />

the product. The choice of the raw materials, plastics and all the additional<br />

equipmentthatcomesintodirectcontactwiththeproductmustbe<br />

performed always keeping in mind that cells, as drug products, cannot be<br />

terminally sterilized. The quality of the materials and reagents utilized is<br />

therefore directly related to the quality and degree of purity of the final<br />

product. Information about the available certification must be gathered for<br />

every component and, for critical materials, audits must be performed to<br />

the manufacturing sites to qualify the supplier.<br />

The protocols used for the cell expansion and processing (if necessary)<br />

must be designed trying to reduce at a minimum the dependence on<br />

growth factors and medium supplements. Each additional component that<br />

is added to the culture medium must be justified and its absence from the<br />

final product must be verified with validated methods. Residues that are<br />

not removed during the production steps must be accurately measured<br />

and limits must be set after performing a risk evaluation analysis, to ensure<br />

that these process-related impurities have no adverse effects on the<br />

patients.<br />

Supplements such as Fetal Bovine Serum (FBS) are permitted for the<br />

manufacturing of cellular therapies [1], as long as the serum is sourced<br />

from a TSE-free area. Anyway, the use of a medium containing FBS should<br />

be limited to the cases for which a valid alternative could not be found.<br />

However, continuous research and development is strongly advised in<br />

order to keep up to date with the latest advances in the field of medium<br />

formulations, in order to be ready to switch to an animal-free medium as<br />

soon as it is feasible. The reduction of growth factors and supplements is<br />

also effective in controlling the manufacturing costs of a cell therapy<br />

product. An evaluation of the economical aspects and market sustainability<br />

should be performed at an early stage to increase the chances for an<br />

industrial development of the cellular product.<br />

The manipulation steps performed during the manufacturing stage should<br />

be kept to a minimum, in order to reduce human intervention and<br />

decrease the risk of contamination. Media fill simulations must be<br />

performed in purposely stressed conditions to ensure that the process and<br />

the facility are able to support the production of a sterile product [2].<br />

When manufacturing patient-specific therapies, extensive efforts should be<br />

directed towards the reduction in the variability of the starting material, that<br />

is usually a tissue sampled from the patient during hospitalization. Working<br />

with well-defined starting material allows for the set-up of a more robust<br />

process with comparable characteristics between batches dedicated to<br />

different patients. The specifications of the final product for parameters such<br />

as cell number, purity and potency must be wide enough to tolerate the<br />

normal biological variability of living organisms, but sufficiently narrowed<br />

down to generate comparable batches of drug. This uniformity is mandatory<br />

for the set up of clinical trials aiming at gathering a reliable analysis of the<br />

safety, tolerability and efficacy data obtained from treated patients, in order<br />

to speed up the clinical development of innovative medicinal products such<br />

as cellular therapies.<br />

References<br />

1. Note for guidance on the use of bovine serum in the manufacture of<br />

human biological medicinal products. , CPMP/BWP/1793/02.<br />

2. Annex 1 - Manufacture of Sterile medicinal Products. EU Guidelines to<br />

Good Manufacturing Practice EudraLex4.<br />

O4<br />

Biosynthetic pathway deflection – a new cell line engineering approach<br />

Hans Henning von Horsten, Thomas Rose, Volker Sandig *<br />

ProBioGen AG, Berlin, Germany<br />

BMC Proceedings 2011, 5(Suppl 8):O4<br />

With increasing information on genome, transcriptome and metabolome<br />

of commonly used production cell lines, engineering becomes an<br />

increasingly popular approach to achieve desired product attributes,<br />

growth behavior and nutrient consumption. Tools range from feeding<br />

intermediate metabolites, overexpression or deregulation of key enzymes<br />

of a pathway to knock-out and RNA silencing. While conceptionally<br />

simple, the latter approaches are either labor intensive or costly to apply<br />

at large scale.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract O4) Overview of GDP-L-Fucose Biosynthesis showing the point of substrate deflection within the fucose de-novo synthesis pathway.<br />

Fucose targeted glycoengineering: Aiming at glycan modulation we<br />

added another principle to this toolbox: enzymatic deflection of a<br />

biochemical pathway. Fucose is synthesized inside the cell from GDPmannose<br />

via short lived intermediates before it is transported to the<br />

Golgi apparatus for attachment to the nascent glycan (Figure 1).<br />

A bacterial enzyme is used to redirect synthesis towards a heterologous<br />

activated hexose that cannot be utilized by the cell resulting in depletion<br />

of the natural pathway (deflecting enzyme, Figure 1). To our surprise,<br />

even lowest level expression of the enzyme completely abolishes fucose<br />

synthesis in stably modified cells.<br />

The approach allows producing antibodies that are devoid of core fucose at<br />

Fc glycans of the CH2 domain [1]. This modification provides higher<br />

flexibility to the Fc-region of IgG1 antibodies and enhances their binding to<br />

the FcgRIIIa receptor of NK cells - the dominating effector cells in antibody<br />

dependent cytotoxicity (ADCC). Consequently, the potency of antibodies<br />

directed against tumor or infected cells is substantially increased.<br />

In contrast to other strategies the approach is easily applied to the starter<br />

cell line of choice and, moreover, allows modification of fully developed<br />

producer cell lines within weeks.<br />

Simultaneous regulation of multiple cellular pathways: Another concept<br />

for clone engineering is based on simultaneous modulation of multiple<br />

cellular processes. We found that the Rho GTPase cdc42 is a highly suitable<br />

effector molecule for this purpose. This pleiotropic modulator dramatically<br />

boosts antibody titers when overexpressed in the cytosol of pharmaceutical<br />

producer clones (Table 1).<br />

Table 1(abstract O4) CDC42-mediated relative mAb-titer<br />

increase over native clone titers. The clones represent<br />

five different products<br />

Titers of Naïve<br />

mAb producing<br />

clones [g/l]<br />

Titers of cdc42engineered<br />

mAb<br />

producing clones [g/l]<br />

Relative Fold<br />

Increase per<br />

modified clone<br />

0,8 1,65 2,06<br />

0,9 2,2 2,4<br />

2,3 3,0 1,3<br />

2,6 4,5 1,73<br />

0,8 1,65 2,06<br />

Page 5 of 181<br />

Reference<br />

1. von Horsten HH, Ogorek C, Blanchard V, Demmler C, Giese C, Winkler K,<br />

Kaup M, Berger M, Jordan I, Sandig V: Production of non-fucosylated<br />

antibodies by co-expression of heterologous GDP-6-deoxy-D-lyxo-4hexulose<br />

reductase. Glycobiology 2010, 20(12):1607-18.<br />

O5<br />

Fluorescence-based tools to improve biopharmaceutical process<br />

development<br />

Tiago M Duarte 1 , Manuel JT Carrondo 1,2 , Paula M Alves 1,2 , Ana P Teixeira 1,2*<br />

1 Instituto de Biologia Experimental e Tecnológica (IBET), Apartado 12, 2781-901<br />

Oeiras, Portugal; 2 Instituto de Tecnologia Química e Biológica – Universidade<br />

Nova de Lisboa (ITQB-UNL), Apartado 127, 2781-901 Oeiras, Portugal<br />

E-mail: anat@itqb.unl.pt<br />

BMC Proceedings 2011, 5(Suppl 8):O5<br />

Background: Process optimization and control are essential to match<br />

biopharmaceutical market demands. Early-steps in the development of new<br />

processes require screening hundreds of transfected clones in order to<br />

select those with the best expression characteristics, and identifying optimal<br />

environmental conditions for these clones to grow and express the protein<br />

of interest. While some culture systems include in situ analysis of cell<br />

density, most often by optical density or capacitance measurements, the<br />

majority of them still rely on off-line, laborious and time-consuming<br />

protocols for recombinant protein analysis. Spectroscopic methods have<br />

been proposed in literature to support bioprocesses development, because<br />

they are non-invasive and provide data on multiple components present in<br />

the culture bulks, thereby increasing information on process performance<br />

[1]. Here, we developed a fluorescence-based method for real-time<br />

monitoring of viable cells and antibody titers in bioreactor cultures, and<br />

extended this strategy for high throughput analysis of cellular productivity<br />

in 96-well plates.<br />

Materials and methods: For high-throughput cellular productivity analysis,<br />

three CHO-K1 cell clones with different IgG 4 monoclonal antibody specific<br />

productivities were grown in 125mL shake flasks. Cultures were sampled<br />

twice a day to assess cellular growth; cell supernatants were stored at -20°C<br />

for further antibody quantification and fluorescence analysis. The<br />

supernatant samples were distributed into black 96-well plates and the<br />

fluorescence maps were collected in a spectrofluorometer (Horiba Jobin<br />

Yvon) connected to a microwell plate reader via optical fibers, placed above


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

the microtiter plate [2]. For real-time monitoring of bioreactor cultures, the<br />

same three clones were grown in 650mL working volume bioreactors.<br />

Fluorescence analysis was performed in situ, with the optical fibres placed<br />

inside a submerged stainless steel probe, incorporating a quartz lens at the<br />

bottom.<br />

Both excitation and emission slit widths were set equal to 5 nm and<br />

integration time was set at 0.5 sec. Fluorescence excitation-emission matrix,<br />

also called fluorescence map, was recorded in the excitation range of 285 -<br />

485 nm, with a step of 20 nm, and in the emission range of 305 - 565 nm,<br />

with steps of 10 nm, giving a total of 176 excitation/emission wavelength<br />

pairs (lex/lem). The acquisition time of each map is approximately 4.5 min.<br />

Further details are described in the literature [2,3].<br />

Results: The applicability of two dimensional (2D) fluorometry was studied<br />

using three CHO cell clones expressing an IgG 4 to monitor viable cell<br />

density and antibody titer in bioreactor cultures and also in 96-well plates,<br />

targeting high throughput screening of cellular productivity. For the<br />

bioreactor application, the fluorescence analysis of the culture bulk was<br />

performed in situ, whereas for the 96-well plate application the cultures<br />

were performed in shake flasks and only the cell supernatant (without<br />

cells) was analyzed in terms of fluorescence. The three cell clones grow at<br />

similar rates and reach similar maximum cell densities but produce<br />

significantly different antibody quantities. Comparing bioreactor to shake<br />

flask cultures, all cell clones reached higher cell densities and lower<br />

antibody titers in the former system (Figure 1A-D).<br />

As a result of cell growth and metabolism, significant changes were<br />

observed in all fluorescent regions. The global relative changes can be seen<br />

in Figure 1E-F, where it is plotted the ratios between fluorescence maps<br />

collected at late culture stages and maps collected after inoculation, in both<br />

Page 6 of 181<br />

culture systems under study. The larger changes occurred in the region of<br />

the cellular cofactors NAD(H)P (maximum at l ex/l em: 365/455nm), which<br />

increased significantly over culture time. In contrast, the fluorescence in the<br />

regions of tryptophan (maximum at lex/lem: 285/365nm) and flavins<br />

(maximum at lex/lem: 465/520) decreased along culture.<br />

As the excitation light or the light emitted by fluorophors can be absorbed<br />

or dispersed by cells or other medium components, the signal that reaches<br />

the detector does not correlate linearly with the fluorophor concentration.<br />

Related to this, no linear correlations could be obtained between individual<br />

l ex/l em pairs and our target bioprocess variables. Therefore, we adopted a<br />

multivariate statistical method, partial least squares, to construct the<br />

regression models linking the variation found in the spectra with the off-line<br />

measurements of the target variables. The dataset from both culture<br />

systems was split in two parts: one for calibration and the other for model<br />

validation. Both variables were predicted with good accuracies in the two<br />

configurations under study (Figure 1A-D). Importantly, the antibody profiles<br />

in the validation data sets could be described with an average error below<br />

7%. This is a very good result, particularly because the validation data sets<br />

include concentrations above the ranges used to calibrate the models,<br />

revealing the extrapolation potential of the developed models.<br />

Conclusions: Both cell density and secreted antibody could be predicted<br />

with good accuracies in culture supernatant samples, demonstrating the<br />

potential of the method to effectively analyze cellular productivity in 96-well<br />

plate format. This methodology allows to by-pass the time-consuming<br />

ELISA assays and thus contributing to shorten early-phases of process<br />

development. The same approach was successfully implemented for in-situ,<br />

real-time monitoring of viable cells and antibody titer in bioreactor cultures,<br />

allowing continuous evaluation of bioprocess performance.<br />

Figure 1(abstract O5) Correlations between predicted and measured cell density (A, B) or antibody titer (C, D), in 96-well plates or bioreactor cultures, as<br />

indicated. Open circles correspond to calibration data points and full circles correspond to validation data points. The optimum number of latent variables<br />

was selected such that the root mean squared error observed in the validation data set was minimum. For cell density, the best model structures are<br />

composed by 6 or 7 latent variables (LVs), corresponding to average errors of 10% or 9%, in the microtiter plates or bioreactor cultures, respectively.<br />

Larger models of 9 LVs allowed the best description for antibody titer, with average errors below 7%. Contour plots show the relative fluorescence<br />

changes in the map during a culture, analyzed in (E) 96-well plates and (F) in bioreactors. Colors reflect the magnitude of the ratio between maps from<br />

the end of a CHO cell culture and the beginning of the same culture.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Acknowledgments<br />

The authors gratefully acknowledge the financial support from Fundação<br />

para a Ciência e a Tecnologia, Portugal (PTDC/EBB-EBI/102750/2008) and<br />

MIT-Portugal Program.<br />

References<br />

1. Teixeira AP, Oliveira R, Alves PM, Carrondo MJT: Advances in on-line<br />

monitoring and control of mammalian cell cultures: Supporting the PAT<br />

initiative. Biotechnol Adv 2009, 27(6):726-732.<br />

2. Teixeira AP, Duarte TM, Oliveira R, Carrondo MJT, Alves PM: Highthroughput<br />

analysis of animal cell cultures using two-dimensional<br />

fluorometry. J. Biotechnol 2011, 151:255-260.<br />

3. Teixeira AP, Duarte TM, Carrondo MJT, Alves PM: Synchronous<br />

fluorescence spectroscopy as a novel tool to enable PAT applications in<br />

bioprocesses. Biotechnol & Bioeng 2011, 108(8):1852-1861.<br />

O6<br />

Towards rational engineering of cells: Recombinant gene expression in<br />

defined chromosomal loci<br />

Kristina Nehlsen 1 , Leonor da Gama-Norton 1,2 , Roland Schucht 1,3 ,<br />

Hansjörg Hauser 1 , Dagmar Wirth 1*<br />

1 Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;<br />

2 Instituto de Tecnologia Química e Biológica-Universidade Nova de Lisboa/<br />

Instituto de Biológica Experimental e Tecnológica (ITQB-UNL/IBET), P-2781-<br />

901 Oeiras, Portugal; 3 InSCREENeX GmbH, 38124 Braunschweig, Germany<br />

E-mail: dagmar.wirth@helmholtz-hzi.de<br />

BMC Proceedings 2011, 5(Suppl 8):O6<br />

The strength of recombinant gene expression is a key property of cell lines<br />

for biopharmaceutical protein production. In most stable cell lines the<br />

expression vector is stably introduced into the host chromosomal DNA.<br />

Apart from the copy number and the used expression control elements the<br />

performance of recombinant expression vectors is modulated by genetic<br />

and epigenetic features provided by flanking host elements. Since targeted<br />

integration is very difficult cell clones with high expression of a recombinant<br />

Page 7 of 181<br />

vector are created by random integration and large scale screening for gene<br />

expression. This allows the isolation of those rare recombinant cells in which<br />

gene expression is optimal. This is usually due to locus-specific influences of<br />

the chromosomal surroundings.<br />

We have developed an efficient methodology for targeting expression<br />

cassettes to specific chromosomal sites [1,2]]. The method (Flp recombinase<br />

mediated cassette exchange - RMCE) allows the repeated use of defined loci<br />

by targeting constructs for expression of proteins and viruses, thereby<br />

allowing to exploit the positive features of a given integration site [3,4]].<br />

Thereby, a systematic evaluation of the performance of a set of expression<br />

vectors in various chromosomal sites becomes feasible.<br />

In this study we screened for high performance integration sites in HEK293<br />

and CHO-K1 cells supporting expression cassettes driven by a potent<br />

promoter. As a read out, production of antibodies and recombinant retroviral<br />

vectors were used. Thereby, we could show that high level expression of a<br />

given promoter is restricted to defined integration sites, while other sites<br />

show only moderate expression (data not shown). An important new finding<br />

was that a given chromosomal site is not flexible with respect to the<br />

integrated cassette but requires the integration of specific promoters. As<br />

illustrated in figure 1, an integration site, identified for supporting high level<br />

expression of an SV40 promoter driven cassette fails to adequately support<br />

expression of MPSV and adenoviral major late promoter (AdmlP). Vice versa,<br />

another site, initially screened for supporting MSCV promoter expression,<br />

could restore expression of the highly homologous MPSV promoter while the<br />

SV40 promoter and the AdmlP promoter give only moderate expression.<br />

Finally, we tested the impact of the orientation of the cassette in a specific<br />

chromosomal site. We found that some integration sites are flexible with<br />

respect to the orientation of the expression cassettes while others support<br />

expression only in one direction (data not shown).<br />

While classical enhancer elements are known to activate promoters largely<br />

independent from the relative position this finding suggests that other cis<br />

acting elements affect the incoming cassettes in an orientation dependent<br />

manner. Together, this shows that not the nature of integration site and the<br />

design of the vector as such define the performance of a producer cell<br />

clone. Rather, the interplay between these components defines the level<br />

Figure 1(abstract O6) Mode of tagging defines the optimal targeting cassette. Two highly potent chromosomal integration sites were screened<br />

upon random integration of an expression cassette driven by the SV40 promoter and the MSCV promoter, respectively. By means of Flp recombinase<br />

mediated cassette exchange, the screening cassette was exchanged for various expression cassettes, thereby integrating expression cassettes driven by<br />

the SV40 promoter, the adenoviral major late promoter (AdmLP) or the MPSV promoter into the same chromosomal site. The expression level upon<br />

targeting the various cassettes was determined and related to the expression level of the tagged cell.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

and stability of expression. Since these interactions cannot be predicted, the<br />

performance of a vector in a given site has to be evaluated empirically.<br />

In order to exploit favourable sets of chromosomal sites and vectors we made<br />

use of bacterial artificial chromosome (BAC) vectors. By recombineering,<br />

expression cassettes were integrated into pre-selected chromosomal sites as<br />

encoded by BAC vectors. These vectors were randomly integrated into cells<br />

by standard transfection protocols. Clones were isolated and evaluated for<br />

expression. As expected, highly reproducible expression characteristics were<br />

found in individual clones (data not shown). In conclusion, the definition of<br />

favorable combinations of specific integration sites and vector design allow<br />

the rational exploitation of given chromosomal sites. For this purpose,<br />

technologies for site specific genetic manipulation of mammalian cells are<br />

essential. This concerns both targeted integration of expression cassettes into<br />

defined loci (such as RMCE or site specific nuclease induced homologous<br />

recombination) or by transduction of large chromosomal domains (as<br />

provided by BAC vectors). These technologies pave the way for predictable<br />

and high expression of biotechnologically relevant products such as<br />

antibodies and recombinant viral vectors.<br />

References<br />

1. Wirth D, Gama-Norton L, Riemer P, Sandhu U, Schucht R, Hauser H: Road<br />

to precision: recombinase-based targeting technologies for genome<br />

engineering. Current opinion in biotechnology 2007, 18(5):411-419.<br />

2. Schucht R, Coroadinha AS, Zanta-Boussif MA, Verhoeyen E, Carrondo MJ,<br />

Hauser H, Wirth D: A new generation of retroviral producer cells:<br />

predictable and stable virus production by Flp-mediated site-specific<br />

integration of retroviral vectors. Mol Ther 2006, 14(2):285-292.<br />

3. Nehlsen K, Schucht R, da Gama-Norton L, Kromer W, Baer A, Cayli A,<br />

Hauser H, Wirth D: Recombinant protein expression by targeting preselected<br />

chromosomal loci. BMC Biotechnol 2009, 9:100.<br />

4. Gama-Norton L, Herrmann S, Schucht R, Coroadinha AS, Low R, Alves PM,<br />

Bartholomae CC, Schmidt M, Baum C, Schambach A, et al: Retroviral vector<br />

performance in defined chromosomal Loci of modular packaging cell<br />

lines. Hum Gene Ther 2010, 21(8):979-991.<br />

O7<br />

Human hair follicle equivalents in vitro for transplantation and<br />

chip-based substance testing<br />

R Horland 1* , G Lindner 1 , I Wagner 1 , B Atac 1 , S Hoffmann 1 , M Gruchow 1 ,<br />

F Sonntag 2 , U Klotzbach 2 , R Lauster 1 , U Marx 1,3<br />

1 TU Berlin, Institute for Biotechnology, Faculty of Process Science and<br />

Engineering, 13355 Berlin, Germany; 2 Fraunhofer IWS Dresden, 01277<br />

Dresden, Germany; 3 TissUse GmbH, 15528 Spreenhagen, Germany<br />

E-mail: reyk.horland@tu-berlin.de<br />

BMC Proceedings 2011, 5(Suppl 8):O7<br />

Introduction: The ability to create an organoid, the smallest functional unit<br />

of an organ, in vitro across many human tissues and organs is the key to<br />

both efficient transplant generation and predictive preclinical testing<br />

regimes. The hair follicle is an organoid that has been much studied based<br />

on its ability to grow quickly and to regenerate after trauma. Replacing hair<br />

Page 8 of 181<br />

lost due to pattern baldness or more severe alopecia, including that induced<br />

by chemotherapy, remains a significant unmet medical need. By carefully<br />

analyzing and recapitulating the growth and differentiation mechanisms of<br />

hair follicle formation, we recreated human hair follicles in tissue culture that<br />

were capable of producing a hair shaft and revealed a striking similarity to<br />

their in vivo counterparts. Extensive molecular and electron microscopy<br />

analysis were used to track the assembly of follicular keratinocytes,<br />

melanocytes and fibroblasts into the final hair shaft producing microfollicle<br />

architecture. The hair follicle generation process was optimized in terms of<br />

efficiency, reproducibility and compliance with regulatory requirements for<br />

later transplantation. In addition, we developed a procedure to integrate the<br />

de novo created human microfollicles into our existing chip-based human<br />

skin equivalents for substance testing. This would allow the evaluation of<br />

the role of hair follicles in dermal substance transport mechanisms for<br />

cosmetic products. Finally, we describe the challenges and opportunities we<br />

are facing for first-in-man transplantation trials.<br />

Material and methods: Mesenchymal, ectodermal and neuro-ectodermal<br />

originated cells from dissected human hair follicles were isolated and<br />

expanded into multiple passages. Dermal papilla fibroblasts have been kept<br />

under low adherent culture conditions resulting in the formation of dermal<br />

papilla-like aggregates. These spheroids underwent extra cellular matrix<br />

protein coating which mimics basal membrane compositions and thereby<br />

retained their inductive properties. In subsequent co-culture procedures<br />

keratinocyte and melanocyte attachment to the spheroids was forced<br />

allowing further follicular development. Ultra-structural examinations by<br />

scanning electron microscopy and immunofluorescence staining of cryosectioned<br />

microfollicles were performed to reveal spatiotemporal<br />

development and to characterize hair follicle like structures. Microfollicles<br />

were further integrated into full skin equivalents by dermal surface<br />

application or by integration through micromanipulation.<br />

Results: The formation of functional neopapillae (dermal papilla condensates)<br />

needs more than 48 hours. At day 7 of the condensation process<br />

theaggregationofcellsismuchmoredenseandtheformationofan<br />

extracellular matrix becomes visible. After the addition of keratinocytes and<br />

melanocytes, the self-organizing microorganoids follow a stringent pattern<br />

of follicular-like formation by generating polarized segments, sheath<br />

formations and the production of a hair shaft – like fiber (Figure 1). Specific<br />

extracellular matrix proteins (e.g. Collagen IV, Chondroitin-4-sulfate, Versican)<br />

and defined mesenchymal and epithelial markers (e.g. Vimentin, CK15) were<br />

expressed. Selected proliferating (Ki67-positive) and apoptotic (TUNELpositive)<br />

keratinocytes were detected in the outer microorganoid layers but<br />

also in the innermost dermal papilla-like aggregate. Microfollicles in skin<br />

equivalents self-organize in specific distance. Stainings show the integration<br />

of microfollicles below the epidermis.<br />

Conclusion: We show that the de novo formation of human microfollicles<br />

in vitro is accompanied by basic hair follicle like characteristics. The<br />

microfollicles can be used to study mesenchymal-epithelial-neuroectodermal<br />

interactions and for the in vitro testing of hair growth-modulating and<br />

pigmentary effects of substances. Hair follicles represent a long-term<br />

reservoir of topically applied substances. As the hair follicle is highly<br />

vascularized, it supports penetration of substances into the skin and further<br />

Figure 1(abstract O7) Microfollicle formation. SEM images taken from developing microfollicles. After adding keratinocytes and melanocytes to the<br />

culture medium a loosen attachment to the neopapilla is seen (A). Polarization of the early aggregate (B). Assembly, orientation and sheath formation (C);<br />

microfollicle with fiber production (D).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

into the bloodstream. Since 2009 all cutaneous resorption experiments of<br />

the cosmetic industry in the EU have to be performed in vitro. In vitro<br />

testing of topically applied substances might therefore be performed with<br />

significantly enhanced validity by the incorporation of a microfollicle into a<br />

dynamic chip-based bioreactor containing a skin equivalent which mimics a<br />

physiological penetration route. With further improvements, the generated<br />

microfollicles embedded in an artificial skin model might also in future be<br />

used as improved implants for treating wound conditions. Eventually<br />

GMP-compliant manufactured human neopapillae might be used as<br />

implants for treating reduced hair conditions in humans.<br />

O8<br />

Chromosome identification and its application in Chinese hamster<br />

ovary cells<br />

Yihua Cao 1 , Shuichi Kimura 2 , Joon-young Park 1 , Miyuki Yamatani 1 ,<br />

Kohsuke Honda 1 , Hisao Ohtake 1 , Takeshi Omasa 1,2*<br />

1 Department of Biotechnology, Graduate School of Engineering, Osaka<br />

University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan; 2 Institute of<br />

Technology and Science, The University of Tokushima, 2-1 Minamijosanjimacho,<br />

Tokushima 770-8506, Japan<br />

E-mail: omasa@bio.tokushima-u.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):O8<br />

Introduction: Chinese hamster ovary (CHO) cells [1] are today a very<br />

important host for the commercial-scale production of protein<br />

pharmaceuticals. Two sub clones of CHO cells, proline-requiring CHO K1 [2]<br />

and the dihydrofolate reductase (DHFR) gene-deficient CHO DG44 [3],<br />

are the most widely used for both scientific research and industrial<br />

applications [4][5,6]. Previously, we constructed a genomic bacterial artificial<br />

chromosome (BAC) library from mouse Dhfr-amplified CHO DR1000L-4N cell<br />

genome, which was provided 5-fold coverage of the CHO cell genome and<br />

analyzed the structure of amplicons of exogenous Dhfr amplification [7]. The<br />

BAC clones of this library could be landmarks for a physical map for CHO<br />

cell genome that are essential to the basic research and industrial<br />

application of CHO cell genome. In this study, we constructed the detail<br />

chromosomal physical map of CHO DG44 cell and investigated the<br />

chromosome rearrangements among CHO K1, DG44, and primary Chinese<br />

hamster cells. Moreover, to determine the effect of the palindrome structure<br />

on Dhfr amplification in CHO cells, we constructed three types of expression<br />

vectors with or without the junction region of the proposed amplicon and<br />

investigated the gene amplification and expression levels in transfected<br />

CHO DG44 cells.<br />

Materials and methods: Cell lines, culture conditions, construction of<br />

vectors and transfection: CHO DG44, CHO K1 and primary Chinese<br />

hamster cells were used in this study. The primary Chinese hamster cells<br />

were isolated from lung tissue of 4 weeks old female Chinese hamster<br />

[7,8]. The structure of the Dhfr amplicon derived from CHO DR1000L-4N<br />

cells constructed from CHO DG44 cells was determined previously [7]. The<br />

structure has a large palindrome structure containing a small inverted<br />

repeat in the junction region. This small inverted repeat originates from<br />

the integrated vector. On the basis of this junction region, three expression<br />

vectors were constructed [8]. The pSV2-dhfr/GFP vector (vector A) was<br />

constructed from original vector [9] and GFP. The pcD-core region (vector<br />

B) was constructed from the core region (junction region containing two<br />

Dhfr copies and one GFP). The pcD-repeat free core region vector (vector<br />

C) was constructed from the repeat free core region (part of the junction<br />

region containing one Dhfr and one GFP). Three constructed plasmids were<br />

transfected into the CHO DG44 cells. In the Dhfr-amplification step, the<br />

transfected cells were cultivated with MTX at various concentrations of 50,<br />

100, 250 and 500 nM.<br />

Fluorescence in situ hybridization using BAC clones as hybridization<br />

probes (BAC-FISH) and construction of CHO physical map: Chromosome<br />

spreads were prepared from exponential-phase cultures and BAC-FISH to<br />

chromosome spreads was carried out as described previously [7]. In brief,<br />

the BAC probes were detected using fluorescein isothiocyanate (FITC)labeled<br />

streptavidin or an anti-DIG-rhodamine antibody. Chromosomes were<br />

counterstained with 4,6,-diamidino-2-phenylindole (DAPI) and observed<br />

under an Axioskop 2 fluorescence microscope. Photographs were taken<br />

with a CCD camera. After image processing was performed, the ImageJ<br />

software (http://rsbweb.nih.gov/ij/) was used to analyze the chromosomal<br />

Page 9 of 181<br />

loci of the BAC clone probes and the positions of the centromere on the<br />

chromosomes, and expressed as FLpter values [10].<br />

Results and discussion: Construction of BAC-based physical map for<br />

Chinese hamster ovary cells: A CHO genomic BAC library consisted of<br />

122,281 clones was constructed in a previous study [7]. Three hundred BAC<br />

clones were randomly selected from this library and mapped onto each<br />

chromosome of CHO DG44 cells by BAC-FISH. The FISH signal location of<br />

BAC clone probes on each chromosome were determined by digital image<br />

analysis and expressed as FLpter values. The 185 BAC clones were also<br />

mapped on the chromosomes of CHO K1 cell line and 94 clones on<br />

primary Chinese hamster chromosomes to investigate the chromosome<br />

rearrangements. The karyotypic comparison between CHO DG44 and<br />

primary Chinese hamster cells was diagrammatically summarized in Fig. 1.<br />

The 20 chromosomes in CHO DG44 cell line were aligned in order of<br />

decreasing size and assigned letters from A to T. The normal Chinese<br />

hamster chromosome number were estimated using BAC-FISH, endsequencing<br />

and previous comparative study between mouse and Chinese<br />

hamster [11]. The chromosomes A, B, C, F, L, N, R and S were derived from<br />

normal Chinese hamster chromosomes without large rearrangements, and<br />

then these chromosomes were conserved between CHO DG44 and K1 cells<br />

(data not shown). This result suggested that these chromosomes were very<br />

stable and essential in CHO cells and supposedly conserved in other CHO<br />

cell lines.<br />

FISH analysis of gene-amplified chromosomal region of transfected<br />

cells [8]: The chromosomal site of integration of a transgene affects its<br />

transcription rate. This phenomenon is called a positive effect and is often<br />

observed in transgenic organisms in which the transcription of an inserted<br />

transgene is affected by the proximity of the transgene to heterochromatin<br />

[12]. To understand the effect of structure of Dhfr amplicon on the<br />

chromosomal site of integration, we performed two-color BAC-FISH analysis<br />

for transgene. In our previous study, we determined that the amplified gene<br />

is integrated in one specific chromosome (chromosome O). The constructed<br />

vectors B and C contain the palindrome structure obtained from the BAC<br />

clone Cg0031N14 located on chromosome O. The summarized results of<br />

integrated chromosomal sites under various MTX concentrations are shown<br />

in Table 1. Interestingly, the transfected cells whose transgene is located at<br />

the same chromosomal position of the BAC clone Cg0160E04 were<br />

abundantly observed in the vector B and C integrations. It is likely that the<br />

chromosomal position of the BAC clone Cg0160E04 on chromosome O is a<br />

hot spot for Dhfr amplification in CHO cells.<br />

In summary, we constructed a BAC-based physical map for CHO DG44 cells<br />

and analyzed genome-wide rearrangements of chromosome among CHO<br />

cells. This BAC-based physical map will greatly facilitate the studies of CHO<br />

cell genome. The BAC clones comprising this physical map could also<br />

provide a genome-wide resource for analysis of chromosome rearrangements,<br />

chromosome structure, comparative genome hybridization, gene<br />

targeting, and functional genomics.<br />

Acknowledgements<br />

The present work is partially supported by grants from the NEDO of Japan,<br />

the Program for the Promotion of Fundamental Studies in Health Sciences<br />

of the NIBIO, and the Grant-in-Aid for Scientific Research of the JSPS. CHO<br />

BAC library was constructed under the collaboration with Professor<br />

S. Asakawa at the University of Tokyo and Professor N. Shimizu at Keio<br />

University.<br />

References<br />

1. Puck TT: Development of the Chinese hamster ovary (CHO) cell for use<br />

in somatic cell genetics. New York: John Wiley 1985.<br />

2. Kao FT, Puck TT: Genetics of somatic mammalian cells, VII. Induction and<br />

isolation of nutritional mutants in Chinese hamster cells. Proc Natl Acad<br />

Sci U S A 1968, 60(4):1275-1281.<br />

3. Urlaub G, Chasin LA: Isolation of Chinese hamster cell mutants deficient<br />

in dihydrofolate reductase activity. Proc Natl Acad Sci U S A 1980,<br />

77(7):4216-4220.<br />

4. Griffin TJ, Seth G, Xie HW, Bandhakavi S, Hu WS: Advancing mammalian<br />

cell culture engineering using genome-scale technologies. Trends<br />

Biotechnol 2007, 25(9):401-408.<br />

5. Wurm FM: Production of recombinant protein therapeutics in cultivated<br />

mammalian cells. Nat Biotechnol 2004, 22(11):1393-1398.<br />

6. Omasa T, Onitsuka M, Kim WD: Cell engineering and cultivation of Chinese<br />

hamster ovary (CHO) cells. Curr Pharm Biotechnol 2010, 11:233-240.<br />

7. Omasa T, Cao Y, Park JY, Takagi Y, Kimura S, Yano H, Honda K, Asakawa S,<br />

Shimizu N, Ohtake H: Bacterial artificial chromosome library for


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Page 10 of 181<br />

Figure 1(abstract O8) The physical map of CHO DG44 cells and karyotypic comparison to primary Chinese hamster cells on the basis of BAC-FISH results.<br />

Homologous region of CHO DG44 to Chinese hamster were colored according to the estimated Chinese hamster chromosomes.<br />

Table 1(abstract O8) Ratios of amplified genes located at same position of BAC clone Cg0160E04 on chromosome O<br />

MTX concentration (nM) Vector A (%) Vector B (%) Vector C (%)<br />

50 0 (0/14) a<br />

0 (0/11) a<br />

0 (0/15) a<br />

100 0 (0/10) a<br />

33.3 (4/12) a<br />

18.2 (2/11) a<br />

250 0 (0/10) a<br />

61.5 (8/13) a<br />

57.1 (8/14) a<br />

500 0 (0/10) a<br />

a, b<br />

70.8 (17/24) 66.7 (6/9) a<br />

a b<br />

(Number of chromosomes detected at same position of BAC clone Cg0160E04 on chromosome O/total number of analyzed chromosomes). Further results<br />

were obtained from the previous study [8].


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

genome-wide analysis of Chinese hamster ovary cells. Biotechnol Bioeng<br />

2009, 104(5):986-994.<br />

8. Park JY, Yamatani M, Wadano S, Takagi Y, Honda K, Omasa T, Ohtake H:<br />

Effects of palindrome structure on Dhfr amplification in Chinese hamster<br />

ovary cells. Process Biochem 2010, 45(12):1845-1851.<br />

9. Yoshikawa T, Nakanishi F, Itami S, Kameoka D, Omasa T, Katakura Y,<br />

Kishimoto M, Suga K: Evaluation of stable and highly productive gene<br />

amplified CHO cell line based on the location of amplified genes.<br />

Cytotechnology 2000, 33(1-3):37-46.<br />

10. Lichter P, Tang CJ, Call K, Hermanson G, Evans GA, Housman D,<br />

Ward DC: High-resolution mapping of human chromosome<br />

11 by in situ hybridization with cosmid clones. Science 1990,<br />

247(4938):64-69.<br />

11. Yang F, O’Brien PC, Ferguson-Smith MA: Comparative chromosome map<br />

of the laboratory mouse and Chinese hamster defined by reciprocal<br />

chromosome painting. Chromosome Res 2000, 8(3):219-227.<br />

12. Wilson C, Bellen HJ, Gehring WJ: Position effects on eukaryotic gene<br />

expression. Annu Rev Cell Biol 1990, 6:679-714.<br />

O9<br />

Development of an automated, multiwell plate based screening system<br />

for suspension cell culture<br />

Sven Markert * , Klaus Joeris<br />

Roche Diagnostics GmbH, Pharma Biotech Production and Development,<br />

Penzberg, Germany<br />

E-mail: Sven.Markert@roche.com<br />

BMC Proceedings 2011, 5(Suppl 8):O9<br />

Introduction: The automation of cell culture processes becomes more<br />

important in the pharmaceutical industry due to compressed timelines<br />

and the need to develop more products more efficiently. This drive to<br />

develop new processes faster and more efficient requires a streamlined<br />

workflow.<br />

Resource intensive approaches like the use of shake flasks limit the<br />

accessible design space for the development of highly productive<br />

processes or the characterization of established processes. Process<br />

Page 11 of 181<br />

automation provides the appropriate tools to address the following key<br />

points:<br />

Increasing experimental throughput ⇨ “Design of Experiments”<br />

using full factorial designs<br />

Increasing process information ⇨ improve process understanding<br />

(“Quality by Design”)<br />

Automate repetitive manual work ⇨ gain efficiency, focus on<br />

high value tasks<br />

Improve reproducibility ⇨ ensure robust processes<br />

A robotic plate handler based system was selected to meet the demands<br />

of a flexible, fast and modular screening system as presented in figure 1.<br />

Results: Scale-up prediction: The comparability of results obtained with<br />

this new multiwell plate based culture system for suspension adapted cell<br />

lines plates and bioreactors had to be verified. It could be shown that 6well<br />

plates were predictive for a scale-up to a 1000 L stirred tank reactor.<br />

The parameter profiles of viable cell density, lactate and antibody<br />

concentration were comparable in multiwell plates and bioreactors (2 L, 10<br />

L and 1000 L). The plates can be used for process scale-up prediction.<br />

Media screening: An automated media blend screening was carried out in<br />

a second experiment highlighting another main area of application. Two<br />

seed trains of a CHO cell line, media blends of two growth media and two<br />

feeding strategies were screened in 120 wells on 6 well plates. A success<br />

rate of 100% enabled the evaluation of all wells in terms of cell growth and<br />

productivity. An increase in viable cell density and product titer of about<br />

20% in comparison to the reference process was achieved.<br />

2 L bioreactor runs were performed to confirm these optimized parameters.<br />

A total of 6 bioreactor runs using the identified best combination of media<br />

blends, seed train and feeding strategy verified the predicted results from<br />

the multiwell plates and showed an increase in productivity of about 15%.<br />

Conclusion and outlook: The developed robotic screening system is<br />

capable of performing a fully automated workflow consisting of incubation,<br />

sampling, feeding and near real-time analytics. The scalability from the mLscale<br />

up to 1000 L scale could be shown. Furthermore, a successful screening<br />

application was carried out and an increase in product concentration could<br />

be achieved. This potential for a process improvement was confirmed in a<br />

bioreactor study. The robotic screening system has therefore been proven to<br />

be a suitable screening tool for process optimization.<br />

Figure 1(abstract O9) Schematic illustration of the developed robotic screening system prototype. Only the core system is shown with a robotic plate<br />

handler as key device connecting shaken cultivation, processing and analytical components.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Ongoing work is focusing on extending the analytical portfolio by<br />

including additional analytical methods and instrumentation. A second<br />

focus area is in the field of process characterization and process<br />

robustness studies.<br />

Acknowledgements: The authors would like to thank Prof. Dr. Thomas<br />

Scheper (Institute for Technical Chemistry, Leibniz University, Hannover,<br />

Germany) for being the doctoral advisor as well as Katrin Möser and<br />

Pawel Linke for their work on the system during their diploma theses.<br />

O10<br />

Utilization of multifrequency permittivity measurements in addition to<br />

biomass monitoring<br />

Christoph Heinrich * , Tim Beckmann * , Heino Büntemeyer, Thomas Noll<br />

Institute of Cell Culture Technology, Bielefeld University, 33615 Bielefeld,<br />

Germany<br />

E-mail: che@zellkult.techfak.uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):O10<br />

Introduction: In recent years measurement of permittivity signal has been<br />

increasingly used for online biomass monitoring of cell cultures. Breweries<br />

use it as an established method for fermenter inoculation and bioprocess<br />

control for instance [1]. In the case of animal cell cultures the correlation<br />

between permittivity and viable cell densities determined offline varies<br />

along cultivation time. Hence, several authors have used the permittivity<br />

signal as an indirect method for measuring oxygen and glutamine<br />

consumption as well as intracellular nucleotide phosphate concentrations<br />

[2,3]. The latest generation of biomass monitoring devices allows parallel<br />

measurement of permittivity at a range of frequencies leading to an<br />

improvement in the correlation between biomass and permittivity and<br />

providing a tool to further explore other aspects related to the<br />

physiological state of the cells.<br />

Material and methods: In this study 10 different cell lines, among them<br />

industrial cell lines as well as cell lines distributed by ATCC, were cultivated.<br />

Cell type specific chemically defined and animal component-free media<br />

(TeutoCell AG) were used. Batch, fed-batch and perfusion bioreactor<br />

cultivations were carried out in controlled benchtop vessels (Sartorius AG or<br />

Applikon Biotechnology). The i-Biomass 465 sensor (FOGALE nanotech) was<br />

used for online multifrequency permittivity monitoring. In addition the<br />

permittivity signal was used to implement a fully automated cell bleed to<br />

maintain a constant viable cell density in a perfusion process. Furthermore, a<br />

fed-batch feed strategy was introduced to keep the substrate concentration<br />

at a certain level. Cell density and viability were determined using a CEDEX<br />

system (Innovatis-Roche AG). Glucose and lactate were measured with an<br />

YSI 2700 Biochemistry Analyzer (YSI Life Sciences). Amino acids were<br />

quantified using an in-house developed HPLC method.<br />

Page 12 of 181<br />

Results: The FOGALE i-Biomass 465 sensor was used to monitor the viable<br />

cell density of different human, CHO and hybridoma cell cultures online.<br />

A good correlation of the permittivity signal and the offline measured viable<br />

cell density for the growth phase was verified (R > 0.99), but pH-shifts and<br />

increased cell aggregation had a negative impact on the correlation. The<br />

linear factor to calculate the viable cell density from the online permittivity<br />

signal varied between 4.5·10 5 cells/(pF/cm) and 12.0 cells/(pF/cm). A clear<br />

relation between cell type (CHO, human or hybridoma) and the linear factor<br />

could not be established from the available data.<br />

Subsequently, the online biomass monitoring system was used to carry out<br />

a 1 L spin-filter perfusion process with constant viable cell density at a<br />

predefined setpoint. The application of a permittivity closed-loop controlled<br />

cell bleed resulted in a steady concentration of 10 7 viable cells/mL during<br />

perfusion, at a dilution rate of 1.0 d -1 . As soon as this threshold was reached,<br />

the cell bleed was automatically started and controlled based on the online<br />

signal of the i-Biomass 465 sensor.<br />

In addition to the correlation with viable cell density, a linear relationship<br />

(R 2 > 0.96) between the online i-Biomass 465 signal and the concentrations<br />

of numerous components, e.g. glucose, lactate, asparagine, glutamine,<br />

tyrosine, threonine, methionine, lysine, phenylalanine, serine, leucine and<br />

isoleucine, was found during the exponential growth phase of CHO-K1 and<br />

CHO DP-12 cultivations. The results indicated that the number of correlating<br />

substrates depended on the used cell line (CHO, human or hybridoma) and<br />

the process strategy (constant pH or pH-shift). Since, the established<br />

substrate correlations were more robust against process variations, they<br />

were investigated as a basis for a closed-loop feeding strategy in fed-batch<br />

cultivations. Compared to a pre-defined feeding schedule or to intermitted<br />

feeding this would have the advantage of avoiding nutrient limitations and<br />

substrate accumulation that might occur due to unexpected high or low cell<br />

growth. Also, feeding would be independent of human surveillance. The<br />

successful application of a completely automated permittivity-controlled<br />

feeding strategy was proved in two fed-batch runs with CHO DP-12 (ATCC<br />

CRL-12445) cells, as shown in Figure 1.<br />

The feeding of both runs was controlled only through the permittivity signal<br />

in order to maintain the asparagine concentration at a certain level.<br />

Asparagine was chosen due to its central role in cell metabolism and to the<br />

fact that it is usually a limiting substrate in CHO DP-12 cultures. These proofof-concept<br />

runs demonstrated that permittivity-based automated feeding<br />

can be a valuable tool for the optimization of fed-batch process parameters,<br />

such as feeding start, flow rate and composition.<br />

Conclusions: For suspension cultures with single cells and high viability a<br />

linear correlation (R 2 ≥ 0.98) of the permittivity signal with the viable cell<br />

density measured offline was obtained, at least for the exponential growth<br />

phase. Based on this correlation, a closed-loop controlled cell bleed was<br />

implemented in a perfusion process in which the permittivity signal was<br />

used to keep the viable cell density at a constant level of 10 7 viable cells/mL.<br />

Figure 1(abstract O10) Total viable cell count, viable cell density and asparagine concentration of the two proof-of-concept CHO DP-12 fed-batch<br />

processes (initial working volume: 1 L; 37°C; 40% DO; pH 7.1).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Furthermore, a linear correlation with the i-Biomass 465 signal was observed<br />

for several substrates independent of the correlation between viable cell<br />

density and permittivity. Based on these results, a closed-loop controlled<br />

feeding was successfully established resulting in a fully automated fed-batch<br />

process.<br />

Acknowledgements: We would like to thank FOGALE nanotech for<br />

providing the i-Biomass 465 system and TeutoCell AG for supplying the<br />

cell culture media and feed solutions.<br />

References<br />

1. Boulton CA, Maryan PS, Loveridge D: The application of a novel biomass<br />

sensor to the control of yeast pitching rate. Proc 22nd Eur Brew Conv<br />

Zurich European Brewing Convention Oxford: Oxford University Press 1989,<br />

653-661.<br />

2. Noll T, Biselli M: Dielectric spectroscopy in the cultivation of suspended<br />

and immobilized hybridoma cells. J Biotechnol 1998, 63:187-198.<br />

3. Ansorge S, Esteban G, Schmid G: Multifrequency permittivity<br />

measurements enable on-line monitoring of changes in intracellular<br />

conductivity due to nutrient limitations during batch cultivations of<br />

CHO cells. Biotechnol Prog 2010, 26:272-283.<br />

O11<br />

ATF4 over-expression increased IgG 1 productivity in Chinese hamster<br />

ovary cells<br />

Ahmad M Haredy 1 , Akitoshi Nishizawa 1 , Kohsuke Honda 1 , Tomoshi Ohya 2 ,<br />

Hisao Ohtake 1 , Takeshi Omasa 1,3*<br />

1 Department of Biotechnology, Graduate School of Engineering, Osaka<br />

University, Suita, Osaka, 565-0871, Japan; 2 Tanabe Mitsubishi Pharma,<br />

Hirakata, Osaka, Japan, 573-1153; 3 Institute of Technology and Science, The<br />

University of Tokushima, Tokushima, 770-8506, Japan<br />

E-mail: omasa@bio.tokushima-u.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):O11<br />

Introduction: The endoplasmic reticulum (ER) is the major organelle of<br />

synthesis of protein and forms a membranous network throughout the cell.<br />

According to Shimizu and Hendershot [1], about one third of the total<br />

proteins produced are synthesized in the ER. The ER lumen possesses a<br />

unique environment for high quality control for protein folding and<br />

assembly. It contains high concentrations of molecular chaperones, folding<br />

enzymes, and ATP, which aid in proper maturation of proteins [2]. However,<br />

if the amount of proteins to be folded exceeds the capacity of the folding<br />

machineries, unfolded proteins will accumulate in the ER and Unfolded<br />

protein response (UPR) will start. UPR aims at regaining the homeostasis<br />

inside the ER by attenuating the protein translation, activating the folding<br />

machineries, degradation of the unfolded proteins, and finally apoptosis.<br />

Activating transcription factor 4 (ATF4) is a central factor in the UPR<br />

pathways which is involved in folding and processing of secretory proteins.<br />

Our previous research showed that ATF4 over-expression is efficient for<br />

increasing the productivity of antithrombin-III [3,4]. In this study, we<br />

investigated if this approach is product-specific or not.<br />

Materials and methods: Cell line and medium: Serum-free adapted<br />

Chinese hamster ovary DP-12-SF (CHO DP-12-SF) cell line (producing human<br />

anti-IL-8 IgG 1) and Dulbecco Modified MEM medium supplemented with<br />

10% dialyzed fetal bovine serum (FBS) and 200nM methotrexate were used<br />

in this study.<br />

Vector construction: The ATF4 expression plasmid was constructed using<br />

CHO ATF4cDNA into KpnI/XbaI site of the pcDNA3.1/Hygro(+) expression<br />

vector (Invitrogen, Carlsbad, CA, USA), designated as pcDNA3.1/Hygro<br />

(+)-ATF4.<br />

Transfection and selection: The pcDNA3.1/Hygro(+)-ATF4 vector was<br />

transfected into CHO DP-12-SF cell line using a TransIT-LT1 transfection<br />

reagent (Mirus bio Madison, WI USA). Single cell clones were obtained<br />

using the limiting dilution method. The transfected cell lines were selected<br />

using 200μg/ml hygromycin.<br />

Chromosomal integration of ATF4: Genomic DNA was isolated after 72 h<br />

of cultivation using DNeasy blood and tissue extraction kit (Qiagen,<br />

Chatsworth, CA, USA). The primers 5`-TAATACGACTCACTATAGGG-3` and 5`-<br />

TAGAAGGCACAGTCGAGG-3` were employed for the amplification of noncoding<br />

region of pcDNA3.1/Hygro(+)-ATF4 for detection of the integration<br />

of the designated plasmid into the CHO chromosome. PCR was performed<br />

using Ex Taq polymerase (Takara Bio, Otsu, Shiga JAPAN) with 100 ng of the<br />

genomic DNA.<br />

Page 13 of 181<br />

Confirmation of ATF4 expression: RNA was isolated after 72 h of<br />

cultivation using RNeasy mini kit (Qiagen). cDNA equivalent to 1μg of the<br />

previously prepared RNA was prepared using omniscript RT kit (Qiagen)<br />

and PCR was performed using the same primers for chromosomal<br />

detection.<br />

Cell and antibody concentrations: Cell concentration was determined<br />

using Coulter Vi-Cell Automated Cell Viability Analyzer (Beckman Coulter,<br />

Inc., Fullerton, CA, USA). Antibody concentration was determined by<br />

sandwich enzyme linked immunosorbent assay (ELISA) [5]. Kinetic<br />

parameters were calculated as described previously [3].<br />

Real time PCR: The quantification of mRNA of heavy and light chains of IgG<br />

was performed by the SYBR Green real-time RT-PCR method (Applied<br />

Biosystems, Foster City, CA, USA) using the StepOnePlus Real-Time PCR<br />

System.<br />

Results and discussion: The gene encoding ATF4 was cloned from CHO-<br />

K1 inserted into the multiple cloning site of pcDNA3.1 vector with<br />

hygromycin cassette as a selection marker. The ATF4 expression vector was<br />

then transfected into CHO DP-12-SF cell line [5], producing humanized anti<br />

IL-8 Immunoglobulin-1 (IgG 1). Twenty six single cell clones were then<br />

established using the limiting dilution method. To examine if pcDNA3.1/<br />

Hygro(+)-ATF4 was integrated into the CHO chromosome or not, PCR<br />

analysis were performed using the primers designed for non-coding region<br />

of the expression vector. Only 5 clones were confirmed with the insert at<br />

1.2 Kb; CHO DP-12-ATF4-3, -9, -10, -12, and-16. Reverse Transcriptase-<br />

Polymerase Chain Reaction analyses revealed that only 3 clones, CHO DP-12-<br />

ATF4-10, -12, and -16, showed positive ATF4 expression. After 144 hours of<br />

cultivation, only clones with confirmed ATF4 expression showed significant<br />

increase in specific IgG 1 production rate ranging from 1.8 to 2.5 times the<br />

parental CHO DP-12-SF cell. Clone DP-12-ATF4-16 that showed the highest<br />

increase in specific productivity with about no change in the specific growth<br />

rate was subjected to further analysis for quantification of mRNA of heavy<br />

and light chains to determine if over-expression affects the transcription of<br />

mRNA or not. The result was found in agreement with our previous research<br />

that over-expression of ATF4 did not significantly change the level of the<br />

product mRNA [4]. It suggested that ATF4 over-expression might improve<br />

the translation and the secretion without affecting the transcription.<br />

References<br />

1. Shimizu Y, Hendershot LM: Organization of the functions and components<br />

of the endoplasmic reticulum. Adv Exp Med Biol 2007, 594:37-46.<br />

2. Gomez E, Powell ML, Bevington A, Herbert TP: A decrease in cellular<br />

energy status stimulates PERK-dependent eIF2alpha phosphorylation<br />

and regulates protein synthesis in pancreatic beta-cells. Biochem J 2008,<br />

410:485-493.<br />

3. Ohya T, Hayashi T, Kiyama E, Nishii H, Miki H, Kobayashi K, Honda K,<br />

Omasa T, Ohtake H: Improved production of recombinant human<br />

antithrombin III in Chinese hamster ovary cells by ATF4 overexpression.<br />

Biotechnol Bioeng 2008, 100:317-324.<br />

4. Omasa T, Takami T, Ohya T, Kiyama E, Hayashi T, Nishii H, Miki H,<br />

Kobayashi K, Honda K, Ohtake H: Overexpression of GADD34 enhances<br />

production of recombinant human antithrombin III in Chinese hamster<br />

ovary cells. J Biosci Bioeng 2008, 106:568-573.<br />

5. Kim WD, Tokunaga M, Ozaki H, Ishibashi T, Honda K, Kajiura H, Fujiyama K,<br />

Asano R, Kumagai I, Omasa T, Ohtake H: Glycosylation pattern of<br />

humanized IgG-like bispecific antibody produced by recombinant CHO<br />

cells. Appl Microbiol Biotechnol 2010, 85:535-542.<br />

O12<br />

Characterization of novel pneumatic mixing for single-use bioreactor<br />

application<br />

Brian Lee 1* , David Fang 2 , Matthew Croughan 3 , Manuel Carrondo 4 ,<br />

Sang-Hoon Paik 5<br />

1 PBS Biotech Inc., Camarillo, California, 93012, USA; 2 Systems QbD, Newbury<br />

Park, California, 91320, USA; 3 Keck Graduate Institute (KGI), Claremont,<br />

California, 91711, USA; 4 Instituto de Biologia Experimental e Tecnologica<br />

(IBET), Lisbon, 2780-157, Portugal; 5 Green Cross Corp., Yongin-Si, KyungGi-Do,<br />

449-070, South Korea<br />

E-mail: blee@pbsbiotech.com<br />

BMC Proceedings 2011, 5(Suppl 8):O12<br />

Background: The novel bioreactor system from PBS Biotech® is the first<br />

with a pneumatic mixing device powered solely by gas buoyancy,


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract O12) 95% mixing time from 2L to<br />

5,000L working volumes in PBS prototype units<br />

2L 10L 50L 250L 1,000L 5,000L<br />

Gas Flow Rate (VVM) 0.07 0.06 0.05 0.04 0.03 0.02<br />

Mixing Time (sec) 20 25 30 38 53 62<br />

eliminating the need for an external mechanical agitator. The patented<br />

design of the Air-Wheel mixing device promotes not only high<br />

tangential fluid flow around the wheel but also efficient radial and axial<br />

flows. The leverage effect of the Air-Wheel mixing device also allows for<br />

lower gassing requirement (v/v) with increasing working volume,<br />

making the power utilization from the gas buoyancy more efficient with<br />

scale.<br />

A series of physical tests were performed to demonstrate that PBS systems<br />

can promote uniform, homogenous mixing over a wide range of working<br />

volumes and can offer a low-shear environment for cell culture. A series of<br />

biological tests were then performed at various evaluation sites to confirm<br />

the physical test results.<br />

Results: Mixing time was calculated in PBS systems ranging from 2L to<br />

5,000L working volume by measuring the change in conductivity readings<br />

from bolus additions of concentrated salt solution. 95% mixing times were<br />

found to be between 20 sec and 62 sec over this range of working<br />

volumes in the PBS systems (Table 1), significantly shorter than reported<br />

values in conventional stirred tank systems.<br />

Shear stress and turbulent kinetic energy dissipation rate (ε, inm 2 /s 3 ) were<br />

calculated from computational fluid dynamics (CFD) modeling on Star<br />

CCM+ software. Lower average shear stress (τ avg, in Pa) on the impeller<br />

and lower average energy dissipation rate (ε, inm 2 /s 3 )intheimpeller<br />

region were found in the PBS system compared to stirred-tank bioreactors<br />

at typical impeller speeds at the respective working volumes. Comparably<br />

low level of shear stress (


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Background: There is a steadily increasing demand for producing higher<br />

yields of biopharmaceuticals as recombinant proteins, and it is<br />

anticipated that this will further expand during the next decades. Among<br />

the most frequent proteins produced are growth factors, monoclonal<br />

antibodies, hormones and blood coagulation factors.<br />

The production of recombinant proteins can be performed in expression<br />

systems derived from bacteria, yeast, plants, insects or mammals.<br />

Prokaryotic cells divide rapidly, making it possible to produce high yields<br />

of the protein at low costs. They are, however, normally not able to<br />

perform post-translational modification of proteins, which, for those of<br />

mammalian origin, is essential to ensure stability, proper folding and<br />

assembly and thus biological activity. Mammalian cells, on the other<br />

hand, confer post-translational modification. Unfortunately their<br />

cultivation is cost expensive and time consuming, they grow slower, are<br />

more sensitive to contamination and produce lower protein yields than<br />

their prokaryotic counterparts. Despite this, 60-70% of all recombinant<br />

proteins used in therapeutics are produced in mammalian cells. Yield<br />

improvement in mammalian systems is currently an area of major<br />

industrial importance [1].<br />

To achieve this, two main strategies have been used: (1) optimising<br />

the components of the vector containing the gene of interest, and<br />

(2) optimising cell growth and selection. Optimisation of the vector by<br />

genetic engineering has mainly focused on increasing the efficiency by<br />

which the gene is transcribed. The concept being that a high level of<br />

transcription would ultimately lead to a higher protein yield due to<br />

increased availability of mRNA for translation. Vector design, the<br />

chromosomal environment of the plasmid integrated in the host genome<br />

and plasmid copy number, are among the parameters that can contribute<br />

to transcription efficiency. An increased level of mRNA coding for any<br />

secreted protein of interest, however, will only be beneficial if the<br />

transcript is correctly transported to the endoplasmic reticulum and then<br />

effectively translated. This area has hitherto been largely neglected.<br />

Page 15 of 181<br />

Efficient mRNA processing: UniTargetingResearch AS is now developing<br />

and commercialising tools to optimise protein synthesis and secretion, by<br />

ensuring that the mRNA encoding the protein of interest is efficiently<br />

processed. This has proved to be heavily dependent on the presence of<br />

specific genetic targeting elements, namely a selected signal sequence<br />

(SS) in combination with appropriate 5' and 3' untranslated regions (5'<br />

and 3'UTRs). Our focus is currently on the SS, which is translated into the<br />

signal peptide (SP).<br />

Earlier we observed a competition between a selected SS and the 3'UTR<br />

in mediating mRNA targeting to distinct classes of polysomes [2]. We<br />

therefore investigated the effect of different SPs derived from<br />

mammalian secretory proteins on the synthesis/secretion of Gaussia<br />

princeps (a marine copepod) luciferase (Gluc) used as a reporter protein<br />

[3]. The results showed that the choice of SP had a major impact on<br />

synthesis/secretion of Gluc in CHO cells. Contrary to what was expected<br />

the SP of albumin was extremely inefficient (


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

dependent on the AA sequence and even a single mutation at a specific<br />

position has a strong effect on protein yield. In order to test whether or<br />

not the total hydrophobicity of the hydrophobic core region (h-region) in<br />

the SP was of importance, hydropathy scores according to Eisenberg<br />

et al. [4] were calculated. From Table 1 it can be seen that there appears<br />

to be no direct correlation between yield and degree of hydrophobicity<br />

of the h-region and thus the latter is not a reliable measure for the<br />

prediction of the efficiency of an individual SP.<br />

The library concept: We thus adopted an alternative approach where<br />

we exploit the plethora of biological data we have accumulated in a<br />

bioinformatics context. A comparison of the success of individual SPs and<br />

an analysis of their AA composition has allowed us make predictions with<br />

respect to which AAs in which positions are likely to have a decisive<br />

influence on protein synthesis/secretion. Based on this we have<br />

developed a tool, termed UTR ® Tailortech, that provides us with the<br />

opportunity of generating rational SP libraries randomised at chosen<br />

positions. In contrast to a traditional non-rational approach which results<br />

in libraries of astronomic proportions not being manageable, with<br />

UTR ® Tailortech the libraries are considerably reduced in size while<br />

simultaneously being enriched for good performers. This increases the<br />

probability of finding “the needle in the haystack”. The tool also<br />

comprises the concept of generating libraries that are re-usable by<br />

constructing so-called “pre-made” libraries. These high-quality libraries<br />

can be linked in a seamless manner to any protein-coding region<br />

contained in any expression vector as outlined in Fig. 1. When combined<br />

with high-throughput screening technology, a tailored SP for any specific<br />

protein (including difficult-to-express proteins) can readily be defined.<br />

Conclusion: From our studies it is evident that there is considerable room<br />

for improvement of production of a recombinant protein by genetically<br />

modifying the SP utilised in a vector. To our knowledge UTR ® Tailortech is<br />

the first attempt to harness this potential in a rational way.<br />

References<br />

1. De Jesus M, Wurm FM: Manufacturing recombinant proteins in kg-ton<br />

quantities using animal cells in bioreactors. Eur J Pharm Biopharm 2011,<br />

78:184-188.<br />

2. Partridge K, Johannessen AJ, Tauler A, Pryme IF, Hesketh JE: Competition<br />

between the signal sequence and a 3'UTR localisation signal during<br />

redirection of beta-globin mRNA to endoplasmic reticulum: implications<br />

for biotechnology. Cytotechnology 1999, 30:37-47.<br />

3. Knappskog S, Ravneberg H, Gjerdrum C, Tröβe C, Stern B, Pryme IF: The<br />

level of synthesis and secretion of Gaussia princeps luciferase in<br />

transfected CHO cells is heavily dependent on the choice of signal<br />

peptide. J Biotechnol 2007, 128:705-715.<br />

Page 16 of 181<br />

4. Eisenberg D, Weiss RM, Terwilliger TC, Wilcox W: Hydrophobic moments in<br />

protein structure. Faraday Symp Chem Soc 1982, 17:109-120.<br />

O14<br />

High-capacity assay to quantify the clonal heterogeneity in potency of<br />

mesenchymal stem cells<br />

Kim C O’Connor 1* , Katie C Russell 1 , Donald G Phinney 2 , Michelle R Lacey 1 ,<br />

Bonnie L Barrilleaux 1 , Kristin E Meyertholen 1<br />

1 Department of Chemical and Biomolecular Engineering, Tulane University,<br />

New Orleans, LA 70118, USA; 2 Scripps Research Institute, Jupiter, FL, 33458,<br />

USA<br />

E-mail: koc@tulane.edu<br />

BMC Proceedings 2011, 5(Suppl 8):O14<br />

Background: The regenerative capacity of mesenchymal stem cells (MSCs)<br />

is contingent on their content of multipotent progenitors [1]. Despite its<br />

importance to the efficacy of MSC therapies, the clonal heterogeneity of<br />

MSCs remains poorly defined. To address this deficiency, the current study<br />

presents a novel high-capacity assay to quantify the clonal heterogeneity<br />

in MSC potency and demonstrates its utility to resolve regenerative<br />

properties as a function of potency. Human bone marrow was the source<br />

of MSCs in this study. The versatility and accessibility of marrow-derived<br />

MSCs make them a standard for many therapeutic applications.<br />

Materials and methods: Primary MSCs were harvested from the iliac crest<br />

bone marrow of healthy adult volunteers and cultured as previously<br />

described [2]. The in vitro assay developed for this study utilizes a 96-well<br />

format to (1) clone fluorescent MSCs stained with CellTracker Green by<br />

limiting dilution, (2) generate matched clonal colonies, (3) differentiate 3<br />

matched colonies per clone to quantify trilineage potential to exhibit adipo-,<br />

chondro- and osteogenesis as a measure of potency, and (4) cryopreserve<br />

the 4th matched colony of each clone in situ in an undifferentiated state for<br />

future use. Clones of known potency were evaluated for their colonyforming<br />

efficiency as a measure of proliferation potential [3]. Expression of<br />

the heterotypic cell adhesion molecule CD146 on the surface of MSC clones<br />

was measured with flow cytometry.<br />

Results: All eight categories of trilineage potential were detected in<br />

human marrow MSCs. Multipotent MSCs had a higher proliferation<br />

potential than lineage-committed MSCs. Tripotent clones formed colonies<br />

with a median efficiency of 50%, as compared with 14% and 1% for biand<br />

unipotent clones, respectively (p < 0.01). Likewise, colonies that<br />

formed from tripotent clones had the largest median diameter. CD146 may<br />

be a biomarker of MSC potency. Histograms of fluorescence intensity from<br />

Figure 1(abstract O13) Seamless insertion of a randomised SS (SS*) from a pre-made library into a recipient vector. Grey boxes indicate special<br />

restriction sites used in the cloning process.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

pooled tripotent clones labeled with anti-CD146 antibody shifted to higher<br />

CD146 expression relative to the parent MSC preparation from which the<br />

clones were generated; whereas, the histograms for parent MSCs and their<br />

unipotent progeny were similar. In particular, the mean fluorescence<br />

intensity of tripotent clones was nearly twice the value for the parent and<br />

unipotent MSCs (p < 0.05).<br />

Conclusions: The research presented here addresses a basic deficiency in<br />

stem cell technology by developing a quantitative and high-capacity assay<br />

to characterize the clonal heterogeneity of MSC potency. The data suggest a<br />

complex hierarchy of lineage commitment in which proliferation potential<br />

and CD146 expression diminish with loss of potency. The capacity of<br />

multipotent MSCs for ex vivo expansion and their differential expression of a<br />

potential potency marker will facilitate rapid production of efficacious MSC<br />

therapies with consistent progenitor content. The assay has numerous basic<br />

research and clinical applications given the importance of heterogeneity to<br />

the therapeutic potential of MSCs.<br />

Acknowledgements: We thank Alan Tucker for his assistance with flow<br />

cytometry, Dina Gaupp for preparing samples for histology, and Prof.<br />

Darwin Prockop for his helpful conversations and suggestions about this<br />

project. This research was sponsored by grants to Prof. O’Connor from the<br />

National Institutes of Health (R03EB007281) and National Science<br />

Foundation (BES0514242).<br />

References<br />

1. Barrilleaux BL, Phinney DG, Prockop DJ, O’Connor KC: Ex vivo engineering<br />

of living tissue with adult stem cells. Tissue Eng 2006, 12:3007-3019.<br />

Figure 1(abstract P1) NovaSeptum 100mL bag and NovaSeptum case configurations.<br />

2. Barrilleaux BL, Phinney DG, Fischer-Valuck BW, Russell KC, Wang G,<br />

Prockop DJ, O’Connor KC: Small-molecule antagonist of macrophage<br />

migration inhibitory factor enhances migratory response of<br />

mesenchymal stem cells to bronchial epithelial cells. Tissue Eng Part A<br />

2009, 15:2335-2346.<br />

3. Russell KC, Lacey MR, Gilliam JK, Tucker HA, Phinney DG, O’Connor KC:<br />

Clonal analysis of proliferation potential of human bone marrow<br />

mesenchymal stem cells as a function of potency. Biotechnol Bioeng 2011,<br />

108:2716-2726, doi:10.1002/bit.23193.<br />

POSTER PRESENTATIONS<br />

Page 17 of 181<br />

P1<br />

Improvement of cell freezing technologies: towards a fully closed<br />

process<br />

Aurore Polès-Lahille * , Brigitte Lafuente, Virginie Perrier, David Balbuena,<br />

Didier Peyret<br />

Merck Serono Biodevelopment, Martillac, France, 33650<br />

E-mail: Aurore.Lahille@merckgroup.com<br />

BMC Proceedings 2011, 5(Suppl 8):P1<br />

Cell culture from vials usually includes an open phase performed under<br />

laminar flow hoods. Even if disposable and small closed systems are<br />

available, at least the first cell culture step after vial thawing, which is a


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

centrifugation, is performed in disposable but open containers. This step is<br />

critical as the risk of contamination is high and it also has a direct impact<br />

on process timelines.<br />

In order to reduce the risk of contamination during thawing, Merck Serono<br />

Biodevelopment studied the possibility of freezing cell banks in bags and<br />

to directly thawing them directly in disposable and closed containers.<br />

The first step of this study was to evaluate if the DMSO present in<br />

freezing media had to be removed just after vial thawing or not. In<br />

general, this cryoprotective organic solvent is removed by centrifugation<br />

to avoid any toxic effect on cells.<br />

So vials from two different mammalian cell lines were thawed, each with<br />

and without a centrifugation step to remove DMSO, or not. Afterwards the<br />

cell amplification was performed in the same way for the two different<br />

thawing procedures. The population doubling level and the viability were<br />

monitored for more than 15 days and there was no significant impact<br />

neither on cell growth nor on viability whether DMSO was removed or not<br />

just after thawing, or not. The important parameter to take into account was<br />

the percentage of DMSO after dilution which had to be less than 0,5%.<br />

In order to reduce the risk of preparing cell banks, Merck Serono<br />

Biodevelopment looked at different disposable closed systems which allow<br />

cell banks to be prepared without laminar flow. It is possible to expand cells<br />

in closed containers such as spinners, shake flasks or rollers with deep tubes<br />

in addition to bags or disposable bioreactors. In order to concentrate cells or<br />

to change the medium from cell amplification to a cell freezing medium a<br />

centrifugation step may be necessary. Single use 500mL centrifuge tubes<br />

with deep tube and air vent are also available. In order to freeze cells, Merck<br />

Serono Biodevelopment evaluated several bags from several suppliers and<br />

chose the NovaSeptum® 100mL bag and the NovaSeptum® Case. The<br />

combination of these 2 elements ensured the integrity of the bag during<br />

freezing at -80°C (Figure 1-2).<br />

Thus 50mL of two different concentrated mammalian cell suspensions<br />

were frozen at -80°C and thawed in disposable and closed containers.<br />

The cells were not centrifuged just after thawing but expanded in order<br />

to seed a production bioreactor. The cells were also maintained in order<br />

to see if there was a long term impact of non-DMSO removal on viability<br />

and cell growth. The population doubling level, the cell viability obtained<br />

in disposable closed cell culture containers in addition to viable cell<br />

density, metabolism, molecule quantity and quality obtained in<br />

bioreactors were monitored. These parameters were compared to the<br />

same process performed with cells coming from vials.<br />

There was no process performance difference obtained between cells frozen<br />

in bags and cells frozen in vials. Cell freezing in bags without DMSO removal<br />

allows a fully closed cell culture and production process to be performed. It<br />

also allows process timelines to be reduced by least 2 weeks.<br />

The study will continue with a focus on bags which are compatible with<br />

nitrogen containers. The quality aspect will also be studied in order to be<br />

able to prepare a full or part of manufacturing of cell banks in bags.<br />

Figure 2(abstract P1) NovaSeptum 100mL bag and NovaSeptum case configurations.<br />

Page 18 of 181<br />

P2<br />

Disposable bioreactors: from process development to production<br />

Aurore Polès-Lahille 1* , Celine Richard 1 , Sandrine Fisch 1 , David Pedelaborde 1 ,<br />

Sandy Gerby 2 , Nora Kadi 1 , Virginie Perrier 1 , Robert Trieau 1 , David Balbuena 1 ,<br />

Laure Valognes 1 , Didier Peyret 1<br />

1 Merck Serono Biodevelopment, Martillac, France, 33650; 2 Ecole Nationale<br />

Supérieure de Technologie des Biomolécules de Bordeaux, Bordeaux, France,<br />

33000<br />

E-mail: Aurore.Lahille@merckgroup.com<br />

BMC Proceedings 2011, 5(Suppl 8):P2<br />

Single-use bioreactors are commonly used for seeding stainless steel<br />

bioreactors or for producing material. The profitability of these<br />

equipments has been well demonstrated on more that decade. But few<br />

data on its scalability have been published.<br />

In 2010-2011, Merck Serono Biodevelopment performed a study in order to<br />

evaluate the performance of several disposable bioreactors. As different<br />

technologies and scales were available, this study compared the<br />

performance of several types of mixing in single-use bioreactors for<br />

process development and pilot scale production. In addition, the<br />

evaluation was performed for both seeding applications and for clinical<br />

material production.<br />

Thus the feature of this study is the comparison of 3 to 50L disposable<br />

bioreactors with internal mixing system or external agitation (Table 1). In<br />

order to compare their performance with traditional bioreactors, gas flow<br />

rate applied in disposable bioreactors was scaled-down from seeding and<br />

production stainless steel bioreactors. Furthermore the ratio working volume<br />

on total volume and the power input applied to the 3L disposable<br />

bioreactor were similar to those in glass bioreactors.<br />

In order to compare the 5 different types of disposable bioreactors, a fedbatch<br />

process producing a highly glycosylated molecule was performed.<br />

The cell growth during the amplification phase was similar reflected by the<br />

doubling time of the cells. It was similar between traditional bioreactors<br />

(21h in 250L stainless steel seeding bioreactor) and disposable bioreactors<br />

(from 17h in the Nucleo and the Orbital to 19h in the STR).<br />

The quality of the molecule together with the molecule titer and the cell<br />

growth were compared in the 5 single use technologies during a production<br />

process. The process performance was also compared in 250L and 1.25kL<br />

bioreactors and to a 3.6L glass development bioreactor. The overall<br />

production process performance was similar in traditional and disposable<br />

bioreactors (Figure 1).<br />

This study was completed by a characterization of liquid/liquid and gas/<br />

liquid transfers inside each disposable bioreactor in order to estimate their<br />

potential in terms of cell culture.<br />

These cells did not require a lot of oxygen. A low k La (0.44h –1 in the<br />

Nucleo) was sufficient to run this process. The air-flow rates applied in the


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P2) Description of the different disposable bioreactors assessed<br />

Name Mobius®<br />

Cellready3L<br />

Supplier Millipore <br />

Nucleo TM<br />

Pierre Guerin<br />

ATMI<br />

orbital shaking bioreactor during the process were defined according to<br />

Merck Serono Biodevelopment and supplier experience.<br />

These flow rates should have been decreased as k La was higher (3.6h- 1 )<br />

than in traditional bioreactors (2.6 h -1 ).<br />

Mixing time was measured in phosphate saline buffer at the maximum<br />

working volume and the agitation speed applied during production<br />

process. Mixing time was measured by injecting concentrated sodium<br />

hydroxide from the top of the bag and from the bottom of the bag.<br />

The Nucleo and the CellReady 3L system have classical probes, while the<br />

Orbital, the RM and the STR bioreactors have optical probes. Optical<br />

probes were more stable because they took a measure every five seconds.<br />

Classical probes were less stable and showed variations of around 0.03 pH<br />

unit even a few minutes after injection in the Nucleo. Nevertheless, mixing<br />

times on all disposable bioreactors (from 15s in CellReady 3L to 100s in<br />

Orbital) were higher than in traditional bioreactors (12s in 3.6L glass<br />

bioreactor to 28s in 1250L stainless steel bioreactor). For the STR, the<br />

mixing time was close to one minute, which is the recommended<br />

maximum for mammalian cell culture. The Orbital and the Nucleo had a<br />

mixing time below two minutes. Howerver, after sodium hydroxide<br />

injection, pH measured below or close to the final pH value. The<br />

disposable bioreactor which has a mixing time closest to traditional<br />

bioreactors is the CellReady 3L.<br />

Cultibag STR TM<br />

Agitation type Marine impeller Paddle 2 x 3-blade segment<br />

impeller<br />

Cultibag RM TM<br />

Sartorius<br />

Move back and<br />

forth<br />

Cultibag Orbital TM<br />

b-test<br />

Orbital shaking<br />

Minimum working volume 1L 8L 10L 5.5L 25L<br />

Maximum working volume 2,5L 20L 50L 20L 50L<br />

Oxygen regulation Micro or marcro Overlay + Overlay + Sparger Overlay only Overlay only<br />

sparger<br />

Sparger<br />

pH regulation with carbon<br />

dioxide<br />

Headspace or sparger Sparger Sparger Headspace Headspace<br />

Temperature regulation Blanket Jacket Jacket Blanket Blanket<br />

Figure 1(abstract P2) Quantity of highly glycosylated molecule produced in different bioreactors.<br />

Page 19 of 181<br />

Finally, an evaluation grid was applied to choose the best disposable<br />

bioreactor. All these comparisons allowed Merck Serono Biodevelopment<br />

to come to a conclusion about disposable bioreactor use and on the<br />

scalability (up and down) of these disposable systems. It also enabled us<br />

to highlight critical points for disposable bioreactor implementation such<br />

as tubing size and length, use of disposable or reusable probes or factory.<br />

P3<br />

Neuroprotective activity of a new erythropoietin formulation with<br />

increased penetration in the central nervous system<br />

Marina Etcheverrigaray 1* , Natalia Ceaglio 1 , Mónica Mattio 1 , Marcos Oggero 1 ,<br />

Ignacio Amadeo 1,2 , Guillermina Forno 1,2 , Norma Perotti 1 , Ricardo Kratje 1<br />

1 Cell Culture Laboratory, School of Biochemistry and Biological Sciences,<br />

Universidad Nacional del Litoral. Ciudad Universitaria – C.C.242 – (S3000ZAA)<br />

Santa Fe, Provincia de Santa Fe, Argentina; 2 Zelltek S.A. Ruta Nacional 168 –<br />

PTLC – (3000) Santa Fe, Provincia de Santa Fe, Argentina<br />

E-mail: marina@fbcb.unl.edu.ar<br />

BMC Proceedings 2011, 5(Suppl 8):P3<br />

Background: Apart from its hematopoietic effect, erythropoietin (EPO) is a<br />

molecule with high neuroprotective potential. However, its prolonged


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

application may cause serious adverse effects due to the erythropoiesis<br />

stimulation. Therefore, an EPO derivative with neuroprotective properties<br />

but low hematopoietic activity, designated as neuropoietin (rhNEPO), was<br />

developed in our lab using an alternative purification process of the<br />

recombinant human erythropoietic counterpart (rhEPO) produced in CHO<br />

cells [1]. The in vitro cytoprotective activity of rhNEPO on neural phenotype<br />

cells and its brain uptake from blood are herein analyzed.<br />

Results: In vitro citoprotective activity of rhNEPO was analyzed on rat<br />

pheochromocytoma cells (PC-12) differentiated to neural phenotype with<br />

neural growth factor (NGF). Apoptosis was triggered by NGF and serum<br />

withdrawal from cell cultures. Thus, nuclear DNA fragmentation was<br />

analyzed by colorimetric TUNEL detection. One-way analysis of variance<br />

was carried out followed by Dunnett´s multiple comparison test.<br />

Probabilities lower than 0.05 were considered significant (p


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

rhNEPO distribution into the nervous system, causing a faster appearance<br />

into the action site. Moreover, the rapid clearance of rhNEPO from plasma<br />

represents an advantage in the treatment of neurological diseases,<br />

because the continuous presence of EPO in blood is the stimulus that<br />

triggers the production of sanguineous cells with the resulting appearance<br />

of adverse side-effects.<br />

Conclusions: The in vitro anti-apoptotic effect of rhNEPO becomes a<br />

remarkable fact to predict its neuroprotective action in the nervous system.<br />

Furthermore, despite its short plasma half-life, rhNEPO appears promptly<br />

within the CSF, being also a further and a significant fact that encourage the<br />

study of neuroepoetin as a potential drug for the treatment of neurological<br />

diseases, in which, a highly neuroprotective activity with low side effects<br />

and a fast blood-to-brain influx are desirables.<br />

References<br />

1. Mattio M, Ceaglio N, Oggero M, Perotti N, Amadeo I, Orozco G, Forno G,<br />

Kratje R, Etcheverrigaray M: Isolation and characterization of a subset of<br />

erythropoietin glycoforms with cytoprotective but minimal<br />

erythropoietic activity. Biotechnol Progr 2011, doi: 10.1002/btpr.633.<br />

2. Egrie JC, Browne JK: Development and characterization of novel<br />

erythropoiesis stimulating protein (NESP). Nephrol Dial Transplant 2001,<br />

16(Suppl 3):3-13.<br />

3. Fukuda MN, Sasaki H, Lopez L, Fukuda M: Survival of recombinant<br />

erythropoietin in the circulation: the role of carbohydrates. Blood 1989,<br />

73:84-89.<br />

P4<br />

New reporter cell clones to determine the biological activity of human<br />

type I interferons<br />

Milagros Bürgi 1 , Claudio Prieto 1 , Marcos Oggero 1 , Mariela Bollati-Fogolín 2 ,<br />

Marina Etcheverrigaray 1 , Ricardo Kratje 1*<br />

1 Cell Culture Laboratory, School of Biochemistry and Biological Sciences,<br />

Universidad Nacional del Litoral. Ciudad Universitaria – C.C. 242 –<br />

(S3000ZAA) Santa Fe, Provincia de Santa Fe, Argentina; 2 Cell Biology Unit,<br />

Institut Pasteur de Montevideo. Mataojo 2020 (11400) Montevideo, Uruguay<br />

E-mail: rkratje@fbcb.unl.edu.ar<br />

BMC Proceedings 2011, 5(Suppl 8):P4<br />

Background: Interferons (IFNs) are potent biologically active proteins<br />

synthesized and secreted by somatic cells of all mammalian species. They<br />

play an important role in the immune response and defence against viruses<br />

because they have antiproliferative, antiviral and immunomodulatory<br />

activities [1]. They are widely used as biopharmaceuticals, so their potency<br />

must be correctly identified. Usually, the biological activity is quantified by a<br />

bioassay based on its capacity to induce an antiviral state in target cells.<br />

Antiviral assays may be subject to high inter- and intra-assay variations<br />

having the drawbacks of using viruses [2]. Therefore, a set a new human cell<br />

lines derived from different tissues were developed using the enhanced<br />

green fluorescent protein (eGFP) gene under the control of type I IFNinducible<br />

Mx2 promoter. In addition, having reporter gene assays derived<br />

from cells of different tissue origins would allow us to design studies aiming<br />

to evaluate how IFNs induce their actions through the human body.<br />

Results: Three human tissue-derived cell lines: A549 (lung cancer cells),<br />

HEp-2 (epidermoid larynx carcinoma cells) and HeLa (cervical adenocarcinoma<br />

cells) were used to develop new reporter gene assays based on<br />

the expression of the eGFP gene under the control of type I IFN-inducible<br />

Mx2 promoter.<br />

Six stably transfected Mx2/eGFP lines (two of each wild type cell line)<br />

were obtained and selected during 10 days with the antibiotic neomycin.<br />

The Mx promoter sequence, cloned upstream to the eGFP, responded<br />

specifically and quantitatively to type I IFNs (IFN-a and IFN-b) usingthe<br />

new reporter cell lines (Figure 1). Therefore, the three cell lines showed<br />

that eGFP percentage (measured by flow cytometry) rose as IFN increased,<br />

reaching maximum values of 25-85% activated cells, depending on the cell<br />

lines and the type of IFNs. No eGFP expression was observed for negative<br />

controls, indicating that there was no basal expression of the reporter<br />

protein.<br />

Two subtypes of IFN-a and IFN-b were assayed: the E. coli-derived IFNs-a<br />

having the amino acid position 23 occupied by the residue Lys (IFN-a2a)<br />

or by the residue Arg (IFN-a2b) and the E.coli-derived IFN-b with the<br />

residue Cys 17 replaced by Ser (IFN-b1b) or the glycosylated CHO cellderived<br />

IFN-b (IFN-b1a). The dose-response relationships corresponding to<br />

both IFN-b subtypes showed higher slopes (43 ± 5 IU/ml) than those of<br />

IFN-a subtypes (23 ± 12 IU/ml). Considering that type-I IFNs act through a<br />

common receptor complex (ifnar) and that mutational analysis revealed<br />

distinct binding sites for these IFNs on ifnar [3], the above mentioned<br />

results give more evidences that differences in IFN a/b recognition may be<br />

associated with cytoplasmic signaling.<br />

Analyzing in more detail the linear dose-response relationships for each<br />

cell line (Table 1), A549 cells showed the lowest detection limit for all IFN<br />

subtypes (0.24 – 0.48 IU/ml). Comparing IFN-a subtypes, no differences in<br />

detection limit were observed using any of the cell lines. Contrarily,<br />

differences in responses were observed when comparing the activity of<br />

IFN-b subtypes in HEp cell lines. Therefore, the standardization of several<br />

assays to measure the potency of IFNs might be carried out using the cell<br />

lines herein documented.<br />

Conclusions: These systems have several advantages when compared to<br />

antiviral activity assays and other reporter gene systems: they can<br />

Figure 1(abstract P4) Reporter gene assays based on the eGFP gene expression after the induction of Mx2 promoter by type I IFNs.<br />

Page 21 of 181<br />

Table 1(abstract P4) Linear dose-response relationships from reporter gene assays employing different human cell<br />

lines<br />

IFN HEp L1D7 HEp L1G5 HeLa C5G6 HeLa C6C3 A549L2A6 A549L2G9<br />

IFN-a2a 1.52 – 390 12.20 – 25,000 7.8 – 1,000 1.95 – 500 0.24 – 16 0.24 – 125<br />

IFN-a2b 0.76 – 390 12.20 – 50,000 7.8 – 1,000 1.95 – 500 0.24 – 4 0.24 – 8<br />

IFN-b1a 0.05 – 200 3.05 – 6,250 3.9 – 250 1.95 – 31.25 0.24 – 32 0.48 – 62<br />

IFN-b1b 3.05 – 200 12.20 – 50,000 3.9 – 1,000 3.90 – 500 0.24 – 32 0.48 – 62


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

determine the potency of type I IFNs using only one cell line; they are very<br />

fast having the specificity of the Mx promoter. Also, they are sensitive and<br />

safe, showing reproducible responses in a dose-dependent manner.<br />

Outstandingly, the main contribution of this work was the development of<br />

alternative reporter systems as suitable candidates to evaluate the way<br />

that IFNs induce their activity in different human tissues.<br />

References<br />

1. Billiau A: Interferon: the pathways of discovery I. Molecular and cellular<br />

aspects. Cytokine Growth Factor Rev 2006, 17(5):381-409.<br />

2. Girad DJ, Fleischaker RJ: A study showing a high degree of<br />

interlaboratory variation in the assay of human interferon. J Biol Stand<br />

1984, 12(3):265-269.<br />

3. Piehler J, Schreiber G: Mutational and structural analysis of the binding<br />

interface between type I interferons and their receptor ifnar2. J Mol Biol<br />

1999, 294(1):223-237.<br />

P5<br />

Analytical techniques for characterization of raw materials in cell<br />

culture media<br />

Chandana Sharma * , Barry Drew, Kevin Head, Rani Pusuluri, Matthew V Caple<br />

Cell Sciences and Development, SAFC, 13804 W 107 th St, Lenexa, KS 66215,<br />

USA<br />

BMC Proceedings 2011, 5(Suppl 8):P5<br />

Figure 1(abstract P5) Analytical RMC process flow.<br />

Page 22 of 181<br />

Background: A prioritized list of 100 “high-risk” raw materials was<br />

developed based on a risk assessment performed within SAFC. This poster<br />

will focus on the analytical screening of certain “high-risk” raw materials<br />

within the prioritized list to identify any variability and critical contaminants<br />

present. In order to achieve this, orthogonal methods were used that<br />

include ultra-high performance liquid chromatography-mass spectrometry<br />

(U-HPLC/MS) for non-volatile polar components and gas chromatographymass<br />

spectrometry (GC/MS) for volatile non-polar materials. Inductively<br />

coupled plasma-optical emission spectrometry (ICP-OES) was also used to<br />

identify any trace metal contamination present. In addition, the solubility of<br />

the raw materials is also tested to identify any variability within a vendor or<br />

between different vendors.<br />

Results: Figure 1 shows the process flow followed within the analytical<br />

RMC initiative. It describes how each raw material is screened and the<br />

strategy for analyzing any possible contaminant.<br />

Conclusions: The orthogonal methods cited to characterize raw materials<br />

have proven to be robust and reliable for the intended purpose. The<br />

solubility experiment described for amino acids illustrates a significant<br />

difference in solubility limits of amino acids and establishes intra- and<br />

inter-vendor variability. This program has helped SAFC get better insight<br />

into their suppliers’ manufacturing processes. This is a long term initiative<br />

within the organization and the most important goal through the<br />

program is to develop a better understanding of raw materials to deliver<br />

superior products of the finest quality.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P5) Summary of solubility findings on multiple lots of three amino acids<br />

Solubility in 100 ml of a neutral buffer<br />

Vendor Lot No. Lysine.HCl (g) Cystine.2HCl (mg) Tyrosine.2Na (mg)<br />

A 1 5.0 12.4 175.1<br />

2 12.5 35.0 185.8<br />

3 10.0 35.2 185.5<br />

B 1 15.0 30.3 185.8<br />

2 12.5 50.8 195.2<br />

3 12.0 48.2 200.7<br />

C 1 60.0 32.0 N/A<br />

2 60.0 35.2 N/A<br />

3 60.0 32.8 N/A<br />

D 1 60.0 N/A N/A<br />

2 60.0 N/A N/A<br />

Acknowledgements: Our sincere thanks to Mr. T. Bower and Mr. S. Jones<br />

of Sigma-Aldrich for running samples on ICP and on LC/MS respectively.<br />

P6<br />

Next-generation sequencing of the CHO cell transcriptome<br />

Jennifer Becker 1* , Christina Timmermann 1 , Tobias Jakobi 2 , Oliver Rupp 2 ,<br />

Rafael Szczepanowski 3 , Matthias Hackl 4 , Alexander Goesmann 2 , Andreas Tauch 3 ,<br />

Nicole Borth 4 , Johannes Grillari 4 , Alfred Pühler 3 , Thomas Noll 1 , Karina Brinkrolf 3<br />

1 Department of Cell Culture Technology, Center of Biotechnology (CeBiTec),<br />

Bielefeld University, 33615 Bielefeld, Germany; 2 Bioinformatics Resource<br />

Facility, Center of Biotechnology (CeBiTec), Bielefeld University, 33615<br />

Bielefeld, Germany; 3 Department of Genome Research and Systems Biology,<br />

Center of Biotechnology (CeBiTec), Bielefeld University, 33615 Bielefeld,<br />

Page 23 of 181<br />

Germany; 4 Department of Biotechnology, University of Natural Resources<br />

and Applied Life Sciences Vienna, 1180 Vienna, Austria<br />

E-mail: jbecker@uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P6<br />

Since 1957 Chinese hamster ovary (CHO) cells are used for in vitro<br />

cultivation as they require assimilable low sustenance [1]. Today, CHO cell<br />

lines represent the most commonly used mammalian expression system<br />

for the production of therapeutic proteins and are considered as the<br />

mammalian equivalent of E. coli in research and biotechnology [2]. The<br />

production of biopharmaceuticals in CHO cells is superior to protein<br />

production in bacteria, because mammalian cell lines procure complex<br />

folding and post-translational modifications like glycosylation. However,<br />

contrary to the increasing importance in biotechnology and industry,<br />

Figure 1(abstract P6) Self-Self Hybridization experiment of the customized CHO microarray. The dye ratio (M-value) is plotted against the adjusted<br />

p-value (student´s t-test, FDR controlled, a=0.05). The dye ratio was calculated using the mean intensity values (A-value) of all probes belonging to one<br />

transcript from four microarray replicates. Different thresholds were used as evidence for quality: p > 0.05, M= - 1< M < 1 (pink); p ≤ 0.05, -1 < M < 1<br />

(dark blue); p ≤ 0.05, M ≤ -1 || M ≥ 1, A < 6 (red); p ≤ 0.05, M ≤ -1 || M ≥ 1, A ≥ 6 (green); p > 0.05, M ≤ -1 || M ≥ 1 (light blue).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

comprehensive genome and transcriptome information of CHO cell lines is<br />

still rare.<br />

In this study, the pyrosequencing technology from 454 Life Sciences and a<br />

novel assembly approach for cDNA sequences were used to achieve a<br />

major step forward towards unraveling the transcriptome of CHO cells.<br />

CHO cDNA samples derived from different CHO cell lines and growth<br />

conditions were used for the generation of 1.84 mill. high quality<br />

sequencing reads with an average read length of 373 nt summing up to<br />

603 Mb data. Assembly of the sequencing data resulted in 41,039<br />

contiguous sequences. These contigs were grouped by the Newbler<br />

software into 36,383 isotigs and 28,039 isogroups.<br />

Taxonomical classification and comparison to the Mus musculus<br />

transcriptome demonstrated the actual quality of the CHO cell line<br />

sequences.<br />

Metabolic pathways of the central carbohydrate metabolism and<br />

biosynthesis routes of sugars used for protein N-glycosylation were<br />

reconstructed from the transcriptome data. All relevant genes representing<br />

major steps in the N-glycosylation pathway and the central metabolism of<br />

CHO cells were detected. Only fructose-1,6-bisphosphatase (3.1.3.11) and<br />

6-phosphogluconolactonase (3.1.1.31) were not identified within the<br />

pentose phosphate pathway.<br />

The newly sequenced CHO cell line transcriptome was the basis for the<br />

design of a customized CHO microarray. Contig sequences were used for<br />

the design of 94.580 probes. The designed probes cover 31,905 splice<br />

variants of CHO transcripts. With a Self-Self Hybridization experiment<br />

(Figure 1) the functionality of the probes was demonstrated. This<br />

experiment was performed with the same RNA as which was used for<br />

sequencing. Half of this RNA was labeled with Cy3, the other half with Cy5.<br />

For this study the dye intensity, the dye ratio and the adjusted p-value<br />

(student´s t-test, FDR controlled, a=0.05) of four microarray replicates were<br />

analyzed. It is expected, that the two labeled RNA samples bind equally to<br />

the probe, if a transcript is expressed (Figure 1: pink, dark blue, red, light<br />

blue). Only the probes for one transcript could be rejected, because of<br />

their dysfunctionality (Figure 1, green).<br />

This CHO microarray is now available for further experiments and will<br />

support transcriptional analysis of CHO cells under process conditions for<br />

cell line and process optimization. It was used already used successfully<br />

for a gene expression study of CHO DP-12 cells cultivated under sodium<br />

butyrate treatment [3].<br />

Acknowledgements: JB, TJ, and JS acknowledge the receipt of a<br />

scholarship from the CLIB Graduate Cluster Industrial Biotechnology. CT is<br />

supported by BIO.NRW of the Cluster Biotechnology of North Rhine<br />

Westphalia. MH is supported by a BOKU DOC grant. NB and JG are<br />

supported by FWF Biotop, JG is supported by GENAU.<br />

References<br />

1. Puck TT, Cieciura SJ, Robinson A: Genetics of somatic mammalian cells. III.<br />

Long-term cultivation of euploid cells from human and animal subjects.<br />

J Exp Med 1958, 108:945-956.<br />

2. Puck TT: Development of the Chinese Hamster Ovary (CHO) Cell.<br />

Molecular Cell Genetics 1985, 1:37-64.<br />

3. Klausing S, et al: Bioreactor cultivation of CHO DP-12 cells under sodium<br />

butyrate treatment and comparative transcriptome analysis with CHO<br />

cDNA microarrays. in press.<br />

P7<br />

A strategy to obtain recombinant cell lines with high expression levels.<br />

Lentiviral vector-mediated transgenesis<br />

Claudio Prieto * , Diego Fontana, Marina Etcheverrigaray, Ricardo Kratje<br />

Cell Culture Laboratory, School of Biochemistry and Biological Sciences,<br />

Universidad Nacional del Litoral. Ciudad Universitaria – C.C.242 – (S3000ZAA)<br />

Santa Fe, Provincia de Santa Fe, Argentina<br />

E-mail: cprieto@fbcb.unl.edu.ar<br />

BMC Proceedings 2011, 5(Suppl 8):P7<br />

Background: The primary goal of any recombinant protein production is<br />

to achieve successful gene transfer and expression in a target cell. There<br />

are two general categories of delivery vehicles/vectors employed in<br />

protein expression protocols. The first category includes the non-viral<br />

vectors, ranging from direct injection of DNA to complexing DNA with<br />

cationc lipds, polylysine, etc. The second category comprises DNA and<br />

RNA viral vectors.<br />

Viruses have evolved specific mechanism to deliver their genetic material<br />

to target cell nuclei. Virus members of family Retroviridae, e.g. retroviruses<br />

and lentiviruses, are among the most widely used viral vectors. The use<br />

of lentiviral vectors has been increasing because the vector system has<br />

attractive features. Lentiviruses have an advantage over retroviruses in<br />

that they can infect both dividing and non-dividing cells and therefore<br />

have attracted much attention regarding the potential as vectors for<br />

gene delivery/therapy. Once integrated into the genome, recombinant<br />

cell lines are selected using different selection mechanisms.<br />

Results: Lentivirus particles were produced by simultaneous cotransfection<br />

of HEK 293T cells with four plasmids. The packaging construct<br />

(pMDLg/pRRE) [1], the VSV-G-expressing construct (pMD.G) [2], the Revexpressing<br />

construct (pRSV-Rev) [1], and the self-inactivating (SIN) lentiviral<br />

vector construct containing the green fluorescent protein (GFP) reporter<br />

gene (pLV-PLK-eGFP). The medium containing lentiviral particles was<br />

collected 48 h after transfection, clarified by centrifugation 10 min at 2000<br />

rpm and then stored at -80°C. To determine viral titers, HEK 293T cells<br />

were seeded at 3 x 10 4 cell/ml in 6-well plates and mantained for 18 h. The<br />

supernatant was replaced with 1 ml of diluted lentiviral particles<br />

supernatant containing pLV-PLK-GFP, followed by incubation overnight.<br />

Then, the supernatants were replaced with fresh medium. The cells were<br />

analized by flow cytometry and the percentage of GFP positive cells were<br />

counted 96 h post transduction. Titer was calculated from the dilutions at<br />

which the percentage of eGFP-positive cells fall within the range of 1-30%<br />

using the following formula [3,4]: Titer (TU/ml) = [F x C/V] x D; where TU/<br />

ml: transduction units/ml, F: frequency of GFP-positive cells, C: total<br />

number of cells in the well at the time of transduction, V: volume of<br />

inoculum in ml, and D: lentivirus dilution.<br />

The viral titer was as high as 4.4 x 10 8 TU/ml.<br />

After that, HEK 293T cells were seeded at a density of 6 x 10 4 cell/ml in 6well<br />

plates and after 24 h the supernatant was replaced by 1 ml medium<br />

containing lentiviral vectors. 96 h post-transduction the cells were<br />

analyzed by flow cytometry (t=0); then, the cells were incubated with the<br />

puromycin selection agent to obtain stable recombinant cell lines. Two<br />

protocols were employed: A-) Single-step selection protocol: the cells were<br />

Table 1(abstract P7) eGFP expression level of different recombinant cell lines according to puromycin concentration<br />

Recombinant cell line Puromycyn (µg/ml) Single-step selection protocol Multistep gradual selection protocol<br />

CL1 1 1.0<br />

Fold increase eGFP expression (X-mean)<br />

1.0<br />

CL5 5 2.0 2.0<br />

CL10 10 2.0 2.0<br />

CL50 50 2.0 2.0<br />

CL100 100 nv 4.5<br />

CL150 150 nv 4.5<br />

CL200 200 nv 6.0<br />

CL250 250 nv nv<br />

nv: non viable cells.<br />

Page 24 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P7) Flow cytometry of different eGFP recombinant cell lines.<br />

incubated with 1, 5, 10, 50, 100, 150, 200 and 250 µg/ml of puromycin in<br />

different plates, and B-) Multistep gradual selection protocol: the cells were<br />

incubated from 1 up to 250 µg/ml of puromycin, but the selection agent<br />

was gradually changed each 7 days on the same plates (Table 1).<br />

Once the cells were resistant to different concentrations of puromycin,<br />

recombinant cell lines were cryopreserved and analyzed by flow<br />

cytometry to compare the expression of eGFP (x-mean). Recombinant cell<br />

lines showed different eGFP expression levels according to puromycin<br />

concentration. The cell line CL200 showed the highest eGFP expression<br />

level (Figure 1).<br />

Conclusions: Employing the gradual selection protocol, it was possible to<br />

maintain the cells in culture condition up to 200 µg/ml puromycin and<br />

achieve higher expression levels of the reporter gene, between 2 and 6<br />

times depending on puromycin concentration. Contrarily, in the singlestep<br />

selection protocol cells cultures were resistant only up to 50 µg/ml<br />

and expression levels of eGFP were lower. Simultaneously, resistant cell<br />

lines were cloned by limit dilution methods and the resulting cell clones<br />

were also analyzed by flow cytometry. The eGFP expression of each clone<br />

was consistent with the ones observed in the respective resistant cell<br />

lines (data not shown). Therefore, with this strategy of recombinant cell<br />

line selection, it was possible to obtain high eGFP producing stable<br />

cell clones without the use of any gene amplification system.<br />

References<br />

1. Dull T, Zufferey R, Kelly M, Mandel RJ, Nguyen M, Trono D, Naldini L: A<br />

third-generation lentivirus vector with a conditional packaging system.<br />

J Virol 1998, 72:8463-8471.<br />

2. Naldini L, Blomer U, Gallay P, Ory D, Mulligan R, Gage FH, Verma IM,<br />

Trono D: In vivo gene delivery and stable transduction of nondividing<br />

cells by a lentiviral vector. Science 1996, 272:263-267.<br />

3. White SM, Renda M, Nam NY, Klimatcheva E, Zhu Y, Fisk J, Halterman M,<br />

Rimel BJ, Federoff H, Pandya S, et al: Lentivirus vectors using human and<br />

simian immunodeficiency virus elements. J Virol 1999, 73:2832-2840.<br />

4. Sastry L, Johnson T, Hobson MJ, Smucker B, Cornetta K: Titering lentiviral<br />

vectors: comparison of DNA, RNA and marker expression methods. Gene<br />

Ther 2002, 9:1155-1162.<br />

P8<br />

In-situ cell density monitoring and apoptosis detection in adherent<br />

Vero cell bioreactor cultures<br />

Emma Petiot 1* , Amal El-Wajgali 1 , Geoffrey Esteban 2 , Cécile Gény 3 ,<br />

Hervé Pinton 3 , Annie Marc 1<br />

1<br />

Laboratoire Réactions et Génie des Procédés, UPR-CNRS 3349, Nancy-<br />

Université, Vandœuvre-lès-Nancy, France; 2 FOGALE nanotech, Nîmes, France;<br />

3<br />

Sanofi pasteur, Marcy L’Etoile, France<br />

E-mail: emma.petiot@nrc-cnrc.ca<br />

BMC Proceedings 2011, 5(Suppl 8):P8<br />

Background: In cell-based processes, and particularly in viral vaccine<br />

production, cell growth and death are strategic informations to obtain for<br />

process monitoring (ie. scale-up, determination of MOI, TOI, and harvest<br />

time). Dielectric spectroscopy is a tool which was increasingly<br />

implemented on cell-culture bioreactors as it presents great potentials,<br />

compared to other methods, for the in-line monitoring of these two<br />

crucial parameters. Considering viral vaccine production, Vero cells are<br />

Page 25 of 181<br />

one of the most employed cell platform. But, due to its adherent<br />

characteristics, few in-line techniques were developed for cell density<br />

monitoring, on the contrary to the ones available for suspension cells. In<br />

addition, it should be underline that no in-line technique exists for<br />

quantification or detection of the mammalian cell death, despite the<br />

importance of this parameter for cell culture processes.<br />

Materials and methods: The Vero cell line employed in this study was<br />

provided by Sanofi Pasteur. Cells were cultivated in serum-free conditions;<br />

either in a reference medium or in a modified medium with alanineglutamine<br />

peptide (Glutamax®) substituted to glutamine. The adhered cell<br />

population was numbered on haemacytometer after crystal violet treatment.<br />

The apoptotic cells, labelled with annexin V, were quantified by flow<br />

cytometry (Guava). The in-line recording of permittivities at different<br />

frequencies and of the characteristic frequency of the cell population, fc,<br />

were performed by a Fogale Biomass system®.<br />

Theoretical background: The application of the permittivity theory to<br />

mammalian cell density quantification was well described in previous<br />

studies [1]. For a basic comprehension it has to be precised that, in this<br />

method, viable cells are considered as miniature condensators, and<br />

permittivity measurement corresponds to the amplitude of the decharge<br />

curve of cell culture suspension. This curve modelling allowed to relate<br />

permittivity to different physical parameters, such as cell size, cell<br />

membrane capacitance or cell intracellular conductivity as described by the<br />

following equations [2]. From these mathematical relations, a parameter that<br />

will be called specific permittivity of the cells (Δε fogale / C) was calculated.<br />

Most of these physical parameters could be potentially impacted by<br />

changes in the cell physiology and especially by modifications induced by<br />

cell death.<br />

Δε =9.r.B.CM /4=3.ð.r 4 .C.CM<br />

fc = si /(2.B.r.C M)<br />

Δε: permittivity, pF.cm -1<br />

fc: characteristic frequency, Hz<br />

B : biovolume (percentage of viable cell volume in the culture suspension)<br />

C : cell concentration, cell.mL -1<br />

r : cell radius, m<br />

C M : membrane capacitance, F. m -2<br />

si : intracellular conductivity, mS.cm -1<br />

Results:<br />

Dielectric properties of adherent Vero cells: A valid correlation between<br />

biovolume and cell concentration was observed for batch cultures<br />

performed in both media, confirming that the Schwan model [3], originally<br />

developed for spherical cells, was also implementable to cells attached on<br />

microcarrier surface. The slope of the correlation between permittivity and<br />

cell concentration highlighted the impact of the medium composition on<br />

the Vero cell dielectric properties. Indeed, in the modified cell culture<br />

medium, the slope was lower suggesting a reduction of specific permittivity,<br />

and so potential changes in cell dielectric properties (C M and si) (Figure 1).<br />

The Fogale Biomass system® also allowed to observe the impact of cell<br />

density on cell morphology. Indeed, for culture in the reference medium, a<br />

linear correlation was observed under 10 x 10 5 cell.mL -1 . Beyond this cell<br />

density, the specific permittivity decreased indicating a decrease of the<br />

biovolume per cells. This was confirmed by microscopic observation at<br />

different time points of the culture, demonstrating a reduction of cell size<br />

due to carrier surface saturation. So, an accurate monitoring of Vero cells<br />

adhered on microcarriers could be realized with Fogale biomass system®,


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

in different culture media and during the different exponential or decline<br />

culture phases.<br />

In-line detection of Vero cell death: The Fogale biomass system® was<br />

also applied to in-line detect Vero cell death. We only focused on apoptosis,<br />

while no necrotic cells were detected in these Vero cell cultures. Dielectric<br />

spectroscopy parameters were plotted and correlated to apoptotic cell<br />

death occurrence. Thus, a drastic increase of cell apoptosis was observed to<br />

be concomitant with fc parameter increase, whatever the culture medium or<br />

the feeding process used (batch / fed-batch). Plotting the characteristic<br />

frequency derivative, dfc/dt, with the apoptotic cell concentration<br />

highlighted that this derivative always became equal to zero when the<br />

apoptosis occurred. As a proof-of-concept, an apoptotic inducer, the<br />

actinomycin D, was added during the exponential cell growth phase of a<br />

batch culture. In that case the fc parameter presented the same behaviour<br />

confirming the relationship between apoptosis physiological modifications<br />

and the physical parameters impacting fc (r, C M, si).<br />

Conclusion: The first major impact of this work was to demonstrate that<br />

no model adaptation was needed to monitor adherent cell concentration<br />

by using permittivity measurements. An accurate monitoring of adhered<br />

Vero cell concentration was obtained with Fogale Biomass system® until<br />

1x10 6 cell.mL -1 . A further development of the method should be to<br />

monitor higher densities of adherent cells. This could be easily achieved by<br />

monitoring the cell size evolution and correcting the changes induced in<br />

the correlations. The second major impact of this work was to demonstrate<br />

the potentials of this method for the risk-mitigation strategies. Indeed, it<br />

was possible to detect a high increase of cell apoptosis in cell culture<br />

performed with reference operating conditions, but also, an abnormal<br />

increase of apoptotic cell concentration artificially induced during the<br />

culture process. This observation validates the fact that detection of<br />

apoptosis occurrence, due to viral infection for example, could be<br />

monitored with this system.<br />

References<br />

1. Ansorge S, Esteban G, Schmid G: Multifrequency permittivity<br />

measurements enable on-line monitoring of changes in intracellular<br />

conductivity due to nutrient limitations during batch cultivations of<br />

CHO cells. Biotechnol Progr 2010, 26:272-283.<br />

Page 26 of 181<br />

Figure 1(abstract P8) Evolution of the relative permittivity, Δε fogale, with the adhered Vero cell concentration and microscopic observations of<br />

microcarriers at different time points of the culture.<br />

2. Markx GH, Davey CL: The dielectric properties of biological cells at<br />

radiofrequencies: Applications in biotechnology. Enzyme Microb Technol<br />

1999, 25:161-171.<br />

3. Schwan HP: Electrical properties of tissue and cell suspensions. Adv Biol<br />

Med Phys 1957, 5:147-208.<br />

P9<br />

Does earlier use of productivity enhancers during cell line selection<br />

lead to the identification of more productive cell lines?<br />

Alison J Porter * , Atul Mohindra, Juana Maria Porter, Andrew J Racher<br />

Lonza Biologics plc, 228 Bath Road, Slough, Berkshire, SL1 4DX, UK<br />

E-mail: alison.porter@lonza.com<br />

BMC Proceedings 2011, 5(Suppl 8):P9<br />

Background: After selection of a recombinant cell line for the production<br />

of a therapeutic protein, attempts are often made to increase volumetric<br />

productivity. Various techniques have been employed during the<br />

production process when trying to increase product concentration. Some<br />

aim to increase the time integral of viable cell concentration (IVC; the<br />

number of hours viable cells are available to produce the product) by<br />

increasing the maximum viable cell concentration and maintaining high<br />

viabilities. Techniques include (i) optimization of media and feeds, (ii)<br />

optimization of feeding strategies and (iii) genetic manipulation. Others<br />

aim to increase specific production rate (Q P) by the deliberate inhibition of<br />

cell growth (controlled proliferation). Three methods commonly used to<br />

control proliferation are use of (i) chemical agents, (ii) hypothermic<br />

conditions and (iii) genetic manipulation. In this study, we focused on<br />

increasing volumetric productivity by manipulating Q P; in particular, by the<br />

use of chemical agents.<br />

As a cell line has typically already been selected as a manufacturing cell<br />

line prior to assessing such methods to increase Q P, the likelihood of<br />

success is not predictable: resulting in the frequently heard comment that<br />

results are ‘cell line specific’.<br />

But what if we were to look at these methods with many cell lines, at a<br />

much earlier stage of development (before the final cell line has been


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P9) Full distribution of product concentration values for the control, NaBu and NaAc cultures.<br />

selected)? It could be that the cell line with the highest product<br />

concentration is typically a non-responder.<br />

The work described looks to answer the questions: (1) To what extent<br />

does the response to such methods vary in a large panel of cell lines<br />

producing the same antibody? and (2) Would their use in an earlier stage<br />

of cell line development enable the selection of a ‘better’ manufacturing<br />

cell line?<br />

Materials and methods: Cell lines: A panel of 148 GS-CHO cell lines was<br />

generated by transfecting the host cell line CHOK1SV with the GS vector<br />

pEE12.4 containing the gene for the model antibody cB72.3 (Porter et al,<br />

2010, Biotechnol Prog 26: 1455-1464).<br />

Cell culture: The cell lines were assessed in a scale-down model (fedbatch<br />

shake-flask cultures) of Lonza Biologics’ final production bioreactor<br />

process, using CDACF medium and feeds. Three cultures were initiated<br />

for each cell line. The first was a control culture, in the second the culture<br />

medium was supplemented with 1 mM Sodium Butyrate (NaBu), and in<br />

the third the culture medium was supplemented with 7.5 mM Sodium<br />

Acetate (NaAc). The cell concentration was determined using a Vi-CELL<br />

automated cell counter. Product concentration was determined using<br />

Protein A HPLC.<br />

Results: The distribution of the values for the parameters IVC, Q P and<br />

product concentration were investigated for each condition (control, NaBu<br />

and NaAc). For IVC, the distribution of the values for both the NaBu and<br />

NaAc conditions are lower than that of the control. For Q P, the distribution<br />

of values for the NaBu condition are higher that that of the control. The<br />

NaAc condition shows no improvement. For product concentration, the<br />

distribution of values for the NaBu condition are lower than that of<br />

the control. Little difference is observed between the control and the NaAc<br />

condition. Data were analyzed by one-way ANOVA and Tukey’s multiple<br />

comparison test at a 5% significance test. The analysis reveals that there is<br />

a significant difference between the control and NaBu conditions for IVC,<br />

QP and product concentration. In addition, there is a significant difference<br />

between the control and NaAc conditions for IVC but not for Q P and<br />

product concentration.<br />

Review of individual cell lines (Figure 1) reveals that<br />

■ The majority of cell lines did not achieve a higher product<br />

concentration compared to the control, when either NaBu or NaAc was<br />

added<br />

■ The use of NaBu resulted in an increase in productivity, compared to<br />

the control, for 27% of the cell lines<br />

■ The use of NaAc resulted in an increase in productivity, compared to<br />

the control, for 41% of the cell lines<br />

■ An increase in productivity was approximately twice as likely to be<br />

observed with the use of NaAc compared to the use of NaBu<br />

Page 27 of 181<br />

■ For both NaBu and NaAC, an increase in product concentration<br />

compared to the control was more likely if the cell line was a low<br />

producer<br />

■ For those cell lines exhibiting an increase in product concentration<br />

compared to the control when NaBu or NaAc was added, 80% and 62%<br />

respectively were in the lower 50% of producers when ranked by control<br />

product concentation<br />

■ The highest product concentration was achieved using control<br />

conditions<br />

■ No advantage of NaAc or NaBu if looking to identify a more productive<br />

cell line from the population<br />

Conclusions: The data generated in the rigorous testing support the<br />

anecdote that the ability of productivity enhancers to increase Q P is ‘cell line<br />

specific’. On average, no increase in mean product concentration was seen<br />

and only ~25-50% cell lines exhibited a benefit. The question ‘Would the<br />

use of productivity enhancers in an earlier stage of cell line development<br />

enable the selection of a ‘better’ manufacturing cell line?’ has been<br />

answered: There is no advantage in their use. If the selection strategy has<br />

identified a high producing cell line, the productivity enhancers are unlikely<br />

to be effective. Also, the highest product concentration was achieved with<br />

control conditions.<br />

Acknowledgements: Lonza Biologics plc, Slough, GB: Cell Culture<br />

Development Groups; Analytical Development Groups.<br />

P10<br />

Quantification of intracellular nucleotide sugars and formulation of a<br />

mathematical model for prediction of their metabolism<br />

Ioscani Jiménez del Val 1* , Judit M Nagy 2 , Cleo Kontoravdi 1<br />

1 Department of Chemical Engineering, Imperial College London, London,<br />

SW7 2AZ, UK; 2 Institute of Biomedical Engineering, Imperial College London,<br />

London, SW7 2AZ, UK<br />

BMC Proceedings 2011, 5(Suppl 8):P10<br />

The US FDA and the European Medicines Agency have recently proposed<br />

the implementation of the Quality by Design (QbD) paradigm to the<br />

manufacture of biopharmaceuticals. Its implementation requires the use of<br />

all available knowledge of a given product, including the parameters that<br />

affect its quality, for the design, optimization and control of the<br />

manufacturing process. The goal is to ensure that quality is built into the<br />

product at every stage of the manufacturing process. Most licensed<br />

monoclonal antibodies (mAbs) are based on the immunoglobulin G isotype<br />

and contain a consensus N-linked glycosylation site on the Cg2 domains of<br />

their heavy chains. Studies have found that the oligosaccharides attached to


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

this site dramatically influence the efficacy of mAbs as therapeutics either by<br />

reducing their serum half-life or by directly affecting the mechanisms they<br />

trigger in vivo[1,2], thus defining glycosylation as a critical quality attribute of<br />

mAbs under the QbD scope. It has been recently proposed that detailed<br />

Page 28 of 181<br />

mathematical models will play a critical role in the design, control and<br />

optimization of biopharmaceutical manufacturing processes under the QbD<br />

scope [3]. To our knowledge, there are currently no mathematical models<br />

that relate mAb glycosylation with cell culture conditions.<br />

Figure 1(abstract P10) Model reproduction of the experimental data. A, B, C and D show reproduction of viable cell counts (Xv), dead cell counts<br />

(Xd), glucose, glutamine, ammonia and product concentrations in conventional culture media (SF-RPMI) and optimized chemically defined media (PF-<br />

BDM). E, F, G and H show reproduction of intracellular NSD concentrations.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Several reports have shown that glycosylation is directly affected by the<br />

intracellular availability of nucleotide sugar donors (NSDs) [4] which are<br />

the co-substrates for the glycosylation reactions that occur in the Golgi<br />

apparatus. During culture, cells synthesize all the relevant NSDs from<br />

glucose through the nucleotide sugar metabolic pathway. In an effort to<br />

relate process conditions with mAb glycosylation, we have generated a<br />

dynamic mathematical model for this metabolic pathway. The NSD<br />

pathway described in KEGG [5] was used as the starting point. In the<br />

full pathway, four potential carbon sources are converted into the eight<br />

main NSDs (UDP-GlcNAc, UDP-Glucose, UDP-Galactose, UDP-GalNAc,<br />

UDP-GlcA, GDP-Man, GDP-Fuc and CMP-Neu5Ac) through 31 enzymatic<br />

reactions. However, many of the intermediary species are difficult to<br />

measure throughout the course of cell culture. For this reason, the<br />

kinetic model was reduced based on the methodology described by<br />

Nolan and Lee [6] whereby sequential reactions along different<br />

branches of the pathway were lumped into single reactions. As an<br />

additional simplification, glucose was considered as the only carbon<br />

source for the pathway. In order to relate NSD metabolism with<br />

macroscopic cell culture variables, a model for cell growth, nutrient<br />

depletion, metabolite accumulation and product secretion was<br />

formulated based on conventional Monod kinetics. Both models were<br />

linked by defining the intracellular glucose accumulation needed for the<br />

NSD model as a function of the glucose maintenance energy term (m s,<br />

glc) from the cell culture model; the outlet of NSDs from the cells was<br />

associated to the product secretion rate.<br />

In order to estimate the unknown parameters of the combined model,<br />

experimental data from Kochanowski and collaborators [7] was used.<br />

First, the parameters from the macroscopic model were estimated from<br />

the data, including the maintenance energy term for glucose. The results<br />

are shown in panels A, B, C and D of Figure 1. Once the cell culture data<br />

was reproduced accurately with the estimated parameters, the unknown<br />

kinetic parameters from the NSD component of the model were<br />

estimated with the intracellular NSD data from Kochanowski et al. [7].<br />

These results are shown in panels E, F, G and H of Figure 1.<br />

Figure 1 shows that, overall, the model reproduces the experimental data<br />

accurately. The only exceptions are the UDP-GlcNAc concentration<br />

profiles for both culture media and the UDP-GalNAc profiles for the SF-<br />

RPMImedium.InthecaseofUDP-GlcNAc,themodelpredictshigher<br />

accumulation of this NSD towards the end of the data set, whereas the<br />

experimental data suggests that the profile flattens out. It is likely that<br />

the model overestimates UDP-GlcNAc accumulation because experimental<br />

data for CMP-Neu5Ac was unavailable and therefore, this NSD was not<br />

considered within the model. From the reduced metabolic network for<br />

NSDs, it is known that CMP-Neu5Ac is produced from UDP-GlcNAc. If<br />

CMP-Neu5Ac is not considered within the model, it is natural that the<br />

model will predict additional accumulation of UDP-GlcNAc. In the case of<br />

UDP-GalNAc for the SF-RPMI medium, overaccumulation for this NSD is<br />

also predicted by the model. It is possible that this is due to the excess<br />

accumulation of UDP-GlcNAc. From the metabolic network, it is known<br />

that UDP-GalNAc is directly synthesized from UDP-GlcNAc. If the model<br />

predicts higher accumulation of the latter, it will certainly predict higher<br />

UDP-GalNAc accumulation as well.<br />

To our knowledge, the model presented in this work is the first to link cell<br />

culture variables with intracellular metabolic processes through the glucose<br />

maintenance energy term (m s,glc). Furthermore, it is the first model to relate<br />

cell culture variables with intracellular NSD concentrations and the results<br />

show that it is capable of reproducing experimental data accurately.<br />

However, in order to achieve better reproduction of experimental data and<br />

obtain higher confidence in the estimated parameters, additional<br />

experimental data is needed. Specifically, the concentration profiles of GDP-<br />

Fuc and CMP-Neu5Ac throughout cell culture would lead to improved<br />

reproduction of experimental data and predictive capabilities of the model.<br />

Furthermore, fed-batch cultures are also necessary to validate the model<br />

and its parameters.<br />

Once validated with additional data, our mathematical model for NSD<br />

metabolism, can be coupled to a model for Golgi N-linked glycosylation.<br />

This combined model would generate a direct link between extracellular<br />

glucose concentration, which is a readily measurable process variable,<br />

and protein glycosylation. Such a combined model has great potential for<br />

the design, control and optimization of manufacturing processes that<br />

produce mAbs with built in glycosylation-associated quality as proposed<br />

under the QbD paradigm.<br />

Acknowledgements: Ioscani Jiménez would like to thank CONACYT and<br />

The Mario Molina Foundation for their financial support. Cleo Kontoravdi<br />

would like to acknowledge the support of Lonza Biologics for her<br />

Fellowship.<br />

In memoriam Dr. Judit M. Nagy.<br />

References<br />

1. Chen X, Liu YD, Flynn GC: The effect of Fc glycan forms on human IgG2<br />

antibody clearance in humans. Glycobiology 2009, 19:240-249.<br />

2. Raju TS: Terminal sugars of Fc glycans influence antibody effector<br />

functions of IgGs. Curr Opin Immunol 2008, 20:471-478.<br />

3. Jimenez del Val I, Kontoravdi C, Nagy JM: Towards the implementation of<br />

quality by design to the production of therapeutic monoclonal<br />

antibodies with desired glycosylation patterns. Biotechnol Prog 2010,<br />

26:1505-1527.<br />

4. Wong DCF, Wong KTK, Goh LT, Heng CK, Yap MGS: Impact of dynamic<br />

online fed-batch strategies on metabolism, productivity and Nglycosylation<br />

quality in CHO cell cultures. Biotechnol Bioeng 2004,<br />

89:164-177.<br />

5. Kanehisa M, Araki M, Goto S, Hattori M, Hirakawa M, Itoh M, Katayama T,<br />

Kawashima S, Okuda S, Tokimatsu T, Yamanishi Y: KEGG for linking<br />

genomes to life and the environment. Nucleic Acids Res 2008, 36:<br />

D480-D484.<br />

6. Nolan R, Lee K: Modeling the dynamics of cellular networks. Methods in<br />

Bioengineering: Systems Analysis of Biological Networks Artech House<br />

Publishers: Jayaraman A, Hahn J 2009.<br />

7. Kochanowski N, Blanchard F, Cacan R, Chirat F, Guedon E, Marc A,<br />

Goergen JL: Influence of intracellular nucleotide and nucleotide sugar<br />

contents on recombinant interferon-g glycosylation during batch and<br />

fed-batch cultures of CHO cells. Biotech Bioeng 2008, 100:721-733.<br />

P11<br />

Design and simulation of a controller system for metabolic shift<br />

regulation in mammalian cells<br />

Damián Baeza 1,3* , Ziomara P Gerdtzen 2,3,4 , Cristian J Salgado 1,2,3<br />

1 Laboratory for Process Modeling and Distributed Computing, Department<br />

of Chemical Engineering and Biotechnology, Faculty of Physical and<br />

Mathematical Sciences, University of Chile, Santiago, Chile; 2 Millennium<br />

Institute for Cell Dynamics and Biotechnology, Department of Chemical<br />

Engineering and Biotechnology, Faculty of Physical and Mathematical<br />

Sciences University of Chile, Santiago, Chile; 3 Department of Chemical<br />

Engineering and Biotechnology, Faculty of Physical and Mathematical<br />

Sciences, University of Chile, Santiago, Chile; 4 Centre for Biochemical<br />

Engineering and Biotechnology, Department of Chemical Engineering and<br />

Biotechnology, Faculty of Physical and Mathematical Sciences University of<br />

Chile, Santiago, Chile<br />

BMC Proceedings 2011, 5(Suppl 8):P11<br />

Background: Experimental evidence shows the existence of multiple<br />

steady states in mammalian cell culture with distinct cellular metabolism<br />

[1]. Different metabolic states are represented by different lactate to<br />

glucose stoichiometric ratios (ΔL/ΔG).AsitisshowninTable1,the<br />

existence of multiple steady states involves the interaction between<br />

metabolic and gene [2].<br />

Changes on the cell culture’s metabolic state have been found to be<br />

related to the amount of residual glucose in a reactor [3]. Experimental<br />

results indicate that cells in a low metabolic state respond immediately to<br />

pulse additions of glucose.<br />

The problem of providing an optimized strategy for glucose feeding in<br />

order to achieve a specific metabolic state is yet to be studied. We<br />

propose a model based strategy for designing a control system for<br />

Table 1(abstract P11) Gene level fold changes<br />

corresponding to low over high ΔL/ΔG states<br />

Page 29 of 181<br />

Enzyme Real Time PCR Microarray cDNA<br />

Lactate Dehydrogenase ↓ 2.4 ↓ 1.9<br />

Pyruvate Kinase ↓ 2.9 ↓ 2.0<br />

Phosphofructokinase ↓ 2.0 ↓ 1.3


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

metabolic state regulation that considers the biological complexity of the<br />

regulation of the cellular system.<br />

Materials and methods: A detailed metabolic model for mammalian cell<br />

metabolism was formulated (complete model); a system of ordinary<br />

differential equations for the main metabolic variables of the following<br />

form:<br />

�<br />

dC � � �<br />

=S⋅r −t(C)<br />

dt<br />

wherein is the metabolite concentration� vector, � S is the stoichiometric<br />

matrix, is the reaction rate vector, and t(C) the transport/consumption<br />

rate vector. The reaction rates can be described by:<br />

r i = ri f (C)<br />

max<br />

�<br />

⋅ i<br />

�<br />

where max<br />

r is the reaction rate constant i and<br />

i f(C) , is a non-linear<br />

i<br />

function that describes the reaction-reactant dependency. Furthermore,<br />

cell growth rate was modeled as the following first order monod function:<br />

m = m<br />

max<br />

e<br />

C glc I<br />

K +C I +C<br />

X<br />

lac<br />

e<br />

X<br />

X<br />

⋅ ⋅<br />

glc<br />

glc<br />

lac<br />

e<br />

lac<br />

The model’s parameters were obtained from literature and through a<br />

fitting process to experimental data; said fitting process was by the least<br />

squares method using the Nelder-Mead simplex method [4].<br />

Once the experimental curves were obtained with the detailed model a<br />

simplified model was traced that includes the main metabolic reactions<br />

in CHO cells, and the same fitting process was carried out. Stability<br />

analysis was carried out on both the detailed and the simplified model in<br />

order to establish the number of feasible steady states for both models.<br />

Enzyme kinetic for lactate dehydrogenase was weighted with an enzyme<br />

factor F, which varies between 0 and 1:<br />

r’ LDH = F ⋅ rLDH<br />

The objective of the same was to associate low ΔL/ΔG with low values of<br />

F. Based on the estimated F values, a Hill activation function that<br />

depends on the residual glucose concentration was adjusted to obtain<br />

the experimental curves of the culture with metabolic shift.<br />

Figure 1(abstract P11) Simulation results for detailed and simplified model.<br />

Results: The results of the curve fitting of the detailed and simplified<br />

models are depicted in Figure 1a and 1b, respectively. However, the<br />

same parameters cannot be used to simulated a cell culture that presents<br />

metabolic shift; the results of the use of the same parameters to simulate<br />

a MAK cell culture induced to a lower metabolic state do not portray the<br />

metabolic response expected for said cell culture (see Figure 1c).<br />

Stability analysis confirmed that both the simplified and detailed<br />

metabolic models exhibit only one steady state. The existence of only<br />

one stable attractor supports the idea that metabolic regulation alone<br />

cannot explain the metabolic shift. Therefore, gene regulation is an<br />

element that should be considered in a model that correctly describes<br />

this phenomenon.<br />

(a) Simulation results for complete model without metabolic shift,<br />

(b) Simulation results for unregulated simplified model without metabolic<br />

shift, (c) Simulation results for unregulated simplified model with<br />

metabolic shift, (d) Simulation results for regulated simplified model with<br />

metabolic shift. - : Simulated Lactate, - : Simulated Glucose, - :<br />

Simulated Cell Conc., � : Lactate, ■ : Glucose, • : Cell Conc.<br />

The implemented regulation model consists of a Hill activation function<br />

that depends on the residual glucose concentration, which modifies the<br />

enzyme kinetic for lactate dehydrogenase in the following manner:<br />

r’ = b ⋅<br />

LDH<br />

n<br />

e<br />

C glc<br />

K + C<br />

X<br />

( )<br />

n<br />

n<br />

e<br />

( glc ) ( glc )<br />

⋅ r<br />

LDH<br />

Page 30 of 181<br />

wherein b = 1 is the maximal expression level, K glc = 1.24 [mM]<br />

is the activation coeficient, n = 14.64 is the Hill coeficient, and C e glc is<br />

the glucose concentration within the bioreactor. The result of<br />

the implementation of the above function is presented in Figure 1d. The<br />

maximal expression level was fixed at 1 due to the upper limit of the<br />

named enzyme activity, the activation coefficient was retrieved from<br />

literature [3] and the Hill coefficient was adjusted through the previously<br />

used curve fitting process. Additionally, the sum of square residuals of<br />

glucose, lactate and cell concentration was 15.74 when comparing the<br />

simplified regulated model with another source of experimental data,<br />

further supporting the proposed metabolic model.<br />

Conclusions: Metabolic shift is caused by regulation at gene expression<br />

and metabolic levels. A reduction of the ΔL/ΔG ratio is accompanied by a<br />

variation in the expression levels of certain genes that participate in<br />

glycolisis, which justifies the use of a regulation function that varies from 0


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

to 1. Stability analysis concludes that the current metabolic models are<br />

capable of reproducing only one steady state, making explicit the need to<br />

implement a regulation model. Moreover, the immediate response<br />

capability to glucose feed pulses indicate that said regulation model must<br />

be continuous. A Hill activation function was for supplying the gene<br />

regulation due to its structure; glucose concentration has been noted<br />

experimentally to be the main factor that produces a metabolic shift in<br />

mammalian cells and the modification of enzyme kinetics leads to a<br />

different ΔL/ΔG ratio, thus to a different steady state.<br />

The final objective of said model is to design a model based controller<br />

capable of maintaining a low metabolic state (ΔL/ΔG ratio under 0.5) under<br />

continuous operation. The tentative input variables are the glucose and<br />

lactate concentrations as well as the ΔL/ΔG ratio. The controller will modify<br />

the response of the system by manipulating the dilution rate and the<br />

glucose feed concentration of the culture.<br />

Acknowledgments: Conicyt, This work has been supported by<br />

FONDECYT Initiation Grants 11080016 and 11090268<br />

References<br />

1. Europa A, Gambhir A, Fu P-F, Hu W-S: Large scale gene expression<br />

profiling of metabolic shift of mammalian cells in culture. Biotechnol<br />

bioeng 2000, 67:25-34.<br />

2. Korke R, Gatti M, Lau A, Lim J, Seow T, Chung M, Hu W-S: Multiple steady<br />

states with distinct cellular metabolism in continuous culture of<br />

mammalian cells. J Biotech 2004, 107:1-17.<br />

3. Cruz HJ, Moreira JL, Carrondo MJT: Metabolic shifts by nutrient<br />

manipulation in continuous cultures of BHK cells. Biotechnol bioeng 1999,<br />

66:104-113.<br />

4. Lagarias JC, Reeds JA, Wright MH, Wright PE: Convergence properties of<br />

the Nelder-Mead simplex method in low dimensions. J Optim 1998,<br />

9:112-147.<br />

P12<br />

The way to a design space for an animal cell culture process according<br />

to Quality by Design (QbD)<br />

Robert Puskeiler 1* , Jan Kreuzmann 1 , Caroline Schuster 1 , Katharina Didzus 2 ,<br />

Nicole Bartsch 1 , Christian Hakemeyer 1 , Heike Schmidt 3 , Melanie Jacobs 3 ,<br />

Stefan Wolf 3<br />

1<br />

Roche Diagnostics GmbH, Pharma Biotech, Development Fermentation,<br />

Penzberg, Germany, 82377; 2 Roche Diagnostics GmbH, Pharma Research and<br />

Early Development Cell Sciences, Penzberg, Germany, 82377;<br />

3<br />

Roche Diagnostics GmbH, Pharma Biotech, Manufacturing Fermentation,<br />

Penzberg, Germany, 82377<br />

E-mail: robert.puskeiler@roche.com<br />

BMC Proceedings 2011, 5(Suppl 8):P12<br />

Background: The strategy of implementation of the QbD (Quality by<br />

design) approach in upstream processing of therapeutic proteins consists<br />

of the identification of critical process parameters (CPPs) that have a<br />

statistically significant influence on the critical quality attributes (CQAs) of<br />

a specific process. By applying the acceptance criteria to the CQAs,<br />

proven acceptable ranges (PARs) for the CPPs can be deduced from<br />

Page 31 of 181<br />

experimental data. The multidimensional combination of these ranges<br />

form the design space and thus assures the quality of the product.<br />

The QbD approach according to the ICH guidelines Q8, Q9 and Q10 may<br />

be subdivided in the work packages scale down model qualification, risk<br />

analysis, process characterization and range studies. The foundation of<br />

the QbD approach is represented by the scale down model. Several<br />

different scale down criteria were applied and adapted until a satisfactory<br />

match of scale down to commercial scale data was achieved. The scale<br />

down model is then used to investigate cause effect relationships<br />

between process parameters and quality attributes of the production<br />

process.<br />

Since a standard cell culture process from thawing of the vial up to the<br />

final production fermenter can comprise up to 100 process parameters, a<br />

risk based approach is helpful to filter the most important ones. Those<br />

parameters are then experimentally investigated to verify their criticality<br />

for the quality attributes of the process. This approach relies on design of<br />

experiment (DoE) to reduce the number of required experiments to a<br />

manageable number while maintaining meaningful results. During the<br />

range studies, those critical parameters will be investigated with the help<br />

of a high resolution DoE matrix in order to be able to reveal possible<br />

interactions and higher order effects.<br />

Scale down model: Based on development data a scale down model at<br />

2 L scale was established. Predefined scale down criteria (power input,<br />

volumetric aeration rate, tip speed) were applied while taking the specific<br />

clone properties into account. The qualification of the scale down model<br />

was carried out by considering an acceptance criteria for several critical<br />

quality attributes and key performance indicators for at least three scale<br />

down fermentation runs. The acceptance criteria consisted of matching<br />

the 2-fold standard deviation range with the mean of the small scale<br />

data and the 3-fold standard deviation range with individual data points.<br />

Risk analysis: Being confronted with a large number of process<br />

parameters, risk assessment tools are used to focus the experimental<br />

efforts on the most relevant parameters. A Failure Mode and Effects<br />

Analysis (FMEA) based tool was applied to rate hypothetical deviations of a<br />

parameter from a previously defined observation range. The hypothetical<br />

deviation is rated by its severity, occurrence and detectability yielding a<br />

ranking of all parameters according to their risk priority number.<br />

Process characterization / range studies: The experimental investigation<br />

was started with a first round fractional factorial screening design with the<br />

parameters filtered by the risk analysis tool. The resulting models were<br />

optimized such that model quality was sufficient (p < 0.05) and no lack of<br />

fit was observed. The parameters were evaluated in terms of statistical<br />

significance by the t-ratio and p-value. Relevance of certain parameters<br />

wascheckedbycomparisonoftheestimatesizetothecenterpoint<br />

variation (Figure 1).<br />

The results of that work package allowed to eliminate some process<br />

parameters from further experimentation due to their lack of significance<br />

and/or relevance. The resulting parameters were investigated in a second<br />

round of multivariate experimentation with a central composite design<br />

aiming at the definition of the unit operation design space. The higher<br />

resolution models were then subsequently used to define the design space<br />

mathematically. To that aim, each quality attribute was evaluated with its<br />

own optimized model only covering significant terms.<br />

Figure 1(abstract P12) Statistical data evaluation from process characterization data. Models with satisfying ANOVA data and lack of fit were created to<br />

evaluate all significant and relevant process parameters. Plots created with JMP®, SAS, Cary, USA. DF = degrees of freedom, C. Total = total sum of<br />

squares, Prob> |t|= probability of getting a higher t-Ratio by chance (p-value).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 2(abstract P12) Example of the visualization of a design space for 4 parameters by the combination of 2 internal and 2 external axes.<br />

Design space visualization: The mathematical definition of the design<br />

space can, for example, be transformed into the following graphical<br />

representation (Fig. 2). Herein, the combination of two external and two<br />

internal parameter axes allow the visualization of 4 parameters in 9 twodimensional<br />

plots. Every single plot depicts the proven acceptable range in<br />

white restricted by the mathematical models represented by the colored<br />

shaded areas. The models are derived from the experimental results and<br />

thus cut those areas that represent exceeded acceptance criteria.<br />

A fully representative view of the 4-dimensional design space can thus be<br />

achieved if all possible permutations of the external and internal axes is<br />

shown.<br />

P13<br />

Improvement of stem cell performance by supplementation with<br />

metabolic enhancers<br />

Abi M Abitorabi 1* , Christopher Wilcox 2<br />

1 2<br />

GIRUS Life Sciences, Inc., Sunnyvale, CA, 94085, USA; Sheffield BioScience,<br />

Beloit, WI, 53511, USA<br />

E-mail: abi@girusinc.com<br />

BMC Proceedings 2011, 5(Suppl 8):P13<br />

Practical culture methods for expansion of human stem cells are needed<br />

for research and industrial applications. In general, ingredients of known<br />

Page 32 of 181<br />

and unknown composition have been used in stem cell cultures based on<br />

past studies and practices. To harness the substantial potential of stem<br />

cells in treating human diseases, products with improved characteristics<br />

are needed to support the manufacturing of stem cells for cell therapy use.<br />

Safety, definition, cost and consistency are key considerations for all new<br />

stem cell products. Based on data mining and cell-based screens, we have<br />

designed the RS Novo defined small molecule metabolic enhancers for<br />

expansion of human embryonic (ESC), mesenchymal (MSC) and<br />

hematopoietic stem cells with limited differentiation. Cell growth, viability,<br />

phenotype and stem cell potential were endpoints of interest in our<br />

studies. Our aim was to minimize media components that would “trigger”<br />

stem cells into differentiation, apoptosis, and necrosis. We also aimed to<br />

minimize undefined components like serum that could introduce<br />

inconsistencies. Here, we cover some RS Novo results with human MSC<br />

and ESC and introduce the GEM Novo serumfreemediumforamore<br />

complete culture system for stem cell expansion. Cells grown in this<br />

culture system maintained their stem cell phenotype and potencies. For<br />

example, at different passages, human ESC H7 clone maintained the Oct-4<br />

marker of undifferentiated cells more consistently in this system versus a<br />

culture system optimized by others (Table 1), and human MSC expanded<br />

in GEM Novo containing RS Novo and then transferred to differentiation<br />

media could differentiate into adipocytes (Figure 1). Altogether, this new<br />

culture system provides a consistent, high performance condition for<br />

human stem cell expansion.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P13) Consistent marker expression at different passages of human ESC culture in GEM Novo medium<br />

containing RS Novo, determined by mean fluorescence intensity (MFI) of Oct-4 staining by flow cytometry<br />

Marker Control Optimized Conditions MFI GEM Novo & RS Novo MFI<br />

Passage 3 Oct-4 75.8 81.5<br />

Passage 8 Oct-4 66.1 81<br />

Figure 1(abstract P13) Human MSC expanded in GEM Novo containing RS Novo can later differentiate into adipocytes as demonstrated here by Oil Red<br />

O staining.<br />

P14<br />

Use of Micro Bioreactor systems to streamline cell line evaluation and<br />

upstream process development for monoclonal antibody production<br />

Steve R C Warr * , Jai Patel, Rongzan Ho, Katy V Newell<br />

GlaxoSmithKline, Stevenage, Hertfordshire, SG1 2NY, UK<br />

BMC Proceedings 2011, 5(Suppl 8):P14<br />

Introduction: The development of monoclonal antibody (mAb) processes<br />

conventionally involves the generation of multiple cell lines in multiwell<br />

plates, cell line screening in shake flasks followed by final cell line selection<br />

and process optimisation in bioreactors. This process can typically take up<br />

to 18 months to generate a robust process suitable for early phase<br />

manufacturing and therefore any opportunity to streamline this process<br />

impacts directly on drug development timelines.<br />

This work describes the potential integration of the Micro-24 Bioreactor<br />

system (Pall Life Sciences) and the Duetz Microflask system (Applikon<br />

Biotechnology) into cell line development and early process development<br />

and demonstrates how these systems can be used for cell line evaluation<br />

and process optimisation. The Micro-24 Bioreactor system comprises 24<br />

bioreactors (7ml working volume) each capable of independent<br />

temperature, dissolved oxygen and pH control. Cell cultures are carried<br />

Page 33 of 181<br />

out in a presterilised polycarbonate mammalian cell culture cassette with<br />

a central vent and are inoculated manually in a laminar flow cabinet<br />

before sealing with Type A single use closures and incubation under<br />

experimental conditions.<br />

Methods: The performance of a number of cell lines in the Micro-24<br />

Bioreactor and the Duetz Microflask system was compared to that in shake<br />

flasks and bioreactors using cell numbers, viability and product titre. This<br />

data was then used to rank cell lines according to specific parameters.<br />

Unless otherwise stated a standard hydrolysate containing complex<br />

medium and standard experimental conditions were used throughout this<br />

work. For shake flasks these were: 35°C, 5% CO 2, 140 rpm; for Duetz<br />

Microflasks: 35°C, 5% CO2, 200 rpm, 80% humidity and for Micro-24<br />

Bioreactors: 35°C, 650rpm, 6.95 pH, 30% DO. Viable cell numbers and<br />

viability were determined using a ViCell Cell Viability Analyser (Beckman<br />

Coulter) and antibody titres were determined using an Immunochemistry<br />

System (Beckman Coulter).<br />

Reproducibility: A hydrolysate containing complex media fed batch<br />

process was run in each of the 24 bioreactors using the same model CHO<br />

cell line. Viable cell numbers (VCC), viability and titre were measured in<br />

each bioreactor and the coefficients of variation (CV) were calculated for<br />

each time point. This data was also used to calculate the specific<br />

productivity (SPR) for each bioreactor. At each time point CVs were


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P14) Comparison of performance rank order of clones in different bioreactor systems in a hydrolysate containing medium batch<br />

process (Figure 1A and B) and fed batch process (Figure 1C, 1D, 1E and 1F).<br />

generally less than 10% for each parameter measured and there was no<br />

significant difference between different rows of the cassette.<br />

Cell Line Selection: To demonstrate the potential of this system to<br />

identify candidate mAb producing cell lines a series of experiments was<br />

carried out using the Micro-24 Bioreactor system and the results<br />

compared to those obtained in our standard cell line selection process.<br />

After initial screening in static multiwell plates during scale up a further<br />

screen in the Duetz Microflask system indicated significant differences<br />

in the performance of the remaining cell lines. In the standard<br />

hydrolysate containing batch process peak titres varied by up to 60%<br />

and there was a 2 fold difference in the overall SPR across the different<br />

cell lines.<br />

Based on titre and SPR data from the Duetz Microflask screen 12 of<br />

these cell lines were selected for further evaluation. Using the standard<br />

batch process conditions these cell lines were grown in parallel in the<br />

Micro-24 Bioreactors, Duetz Microflasks and conventional shake flasks.<br />

Although there were some differences in absolute titres the rank order<br />

of cell lines was similar in each of the systems tested here (Figure 1a<br />

and b) with R 2 =0.66 (Micro-24 v Duetz) and R 2 =0.72 (Micro-24 v shake<br />

flasks).The rank order of peak VCC was also similar in the Micro-24 and<br />

shake flasks (R 2 = 0.7) although there was less similarity in overall SPR<br />

(R 2 =0.4).<br />

This data from the batch process was used to select 6 cell lines for<br />

evaluation in the standard fed batch process which was run in parallel in<br />

the Micro-24 Bioreactor and shake flasks.<br />

For each cell line the effect of feeding was similar in Micro-24 Bioreactors<br />

to shake flasks (Figure 1C) and the rank orders of titre, VCC and SPR<br />

achieved in the Micro-24 Bioreactors were similar to those achieved in<br />

shake flasks (Figure 1D, E and F).<br />

Bioreactor validation: Clones ranked 1, 2, 5 and 12 in the Micro-24<br />

Bioreactors were tested in conventional 2 litre bioreactors. In bioreactors<br />

the titre produced by the top ranked clone was approximately 20% higher<br />

than the clone ranked 12. The remaining 2 clones (ranked 2 and 5 in the<br />

Micro-24) produced intermediate titres.<br />

Product quality: SEC, CE-IEF and NGHC data for the mAb produced in<br />

the Micro-24 showed no significant differences to that produced in<br />

control shake flasks.<br />

Process optimization: The Micro-24 Bioreactor also enables early stage<br />

process optimisation to be carried out at a small scale. The potential for<br />

media development is shown by the data in Table 1A which demonstrates<br />

that the same trend in peak titres is observed when cells are grown in 4<br />

different media in shake flasks, conventional bioreactors and the Micro-24.<br />

Similarly a further experiment in the Micro-24 Bioreactor to investigate the<br />

effect of pH and temperature stepdown on performance demonstrated<br />

Table 1(abstract P14) Process optimization<br />

Table 1A – Medium Development<br />

Medium Shake flasks (120mL) Bioreactor (2000mL) Micro-24 (7mL)<br />

Medium 1 100% 100% 100%<br />

Medium 2 129% 125% 124%<br />

Medium 3 121% 131% 140%<br />

Medium 4<br />

Table 1B – Condition Optimisation<br />

153% 175% 159%<br />

pH Temperature Relative titre<br />

Condition 1 High Low 58%<br />

Condition 2 High High 100%<br />

Condition 3 Low Low 96%<br />

Condition 4 Low High 141%<br />

Page 34 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

that condition dependent titre improvements could be identified using this<br />

instrument (Table 1B).<br />

Discussion: This data demonstrates that the Micro-24 Bioreactor can be<br />

used successfully to select high producing cell lines and to carry out<br />

initial process optimisation experiments. Although there were some<br />

differences in absolute data the rank orders and process trends identified<br />

in the Micro-24 Bioreactors were similar to those in conventional systems.<br />

Therefore this work has shown that the Micro-24 Bioreactor system could<br />

be used to replace shake flasks and bioreactors in cell line evaluation and<br />

early process improvement work.<br />

P15<br />

Cell line selection using the Duetz Microflask system<br />

Steve R C Warr * , Sharon L White, Yuen-Ting Chim, Jai Patel, Hella Bosteels<br />

GlaxoSmithKline, Stevenage, Hertfordshire, SG1 2NY, UK<br />

BMC Proceedings 2011, 5(Suppl 8):P15<br />

Introduction: The identification of a small number of monoclonal<br />

antibody (mAb) producing candidate cell lines from the large number of<br />

clones generated post transfection is one of the bottlenecks of cell line<br />

development. Clone numbers are reduced significantly during initial<br />

medium exchange and static scale up stages but significant numbers can<br />

still progress to evaluation in shaking cultures. This is often carried out in<br />

shake flasks where the number of clones that can be evaluated may be<br />

restricted due to resource limitations.<br />

The Duetz Microflask system is a microtitre plate based system which uses<br />

‘Sandwich Covers’ to convert the individual wells of 24 well plates into<br />

individual ‘mini reactors’. The‘Sandwich Covers’ consist of a stainless steel<br />

Page 35 of 181<br />

cover, a 0.2µm filter, microfibre inlays and a flexible silicone sealing layer<br />

to ensure adequate oxygen transfer rates to individual wells.<br />

This work describes the use of the Duetz Microflask system to evaluate cell<br />

lines in culture prior to scale up to production shake flasks. We have<br />

compared the growth and rank order of performance for a number of cell<br />

lines in the Duetz Microflask system with data obtained from shake flasks<br />

and demonstrated this system can be used successfully to identify<br />

candidate cell lines for further progression.<br />

A series of preliminary experiments was completed to determine suitable<br />

volumes and operating conditions to minimise evaporation. Unless<br />

otherwise stated these were: Fill volume – 1.5mL, Shaker speed – 200rpm,<br />

Humidity – 80%.<br />

Cell line evaluation: After initial transfection a typical cell line development<br />

process would involve plating cell lines to multiwell plates followed by<br />

several static media exchange, scale up and selection steps. These are used<br />

to reduce the number of cell lines carried forward to larger scale static<br />

T-flasks and eventually to shake flasks for further evaluation and selection.<br />

Although the multiwell plate steps are amenable to automation the manual<br />

handling aspects of culturing multiple cell lines in shake flasks can be<br />

resource limiting and therefore the ability to improve cell line selection using<br />

shaking multiwell plates offers significant time line and resource advantages.<br />

Complex medium process: The performance of a series of 28 mAb<br />

producing clones in hydrolysate containing media in Duetz Microflasks<br />

was compared with standard shake flask evaluation data.<br />

The rank order of titre performance of these clones in ‘Early’ Duetz<br />

Microflasks (ie inoculated before repeated shake flask subculture) was<br />

compared to that in shake flasks inoculated after the cultures had been<br />

subcultured for 6 passages. There were some differences in the rank orders<br />

resulting in a correlation R 2 = 0.46. However this improved significantly<br />

Figure 1(abstract P15) Titre rank order of clones in batch shake flasks compared with top clones in batch ‘Early’ ‘Late’ Duetz Microflasks (Figure 1A).<br />

Correlation of the % feed response (Figure 1B) and titre rank order (Figure 1C) of clones in a fed batch process in shake flasks and ‘Late’ Duetz<br />

Microflasks is also shown.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

(R 2 = 0.92) when a further set of Duetz Microflasks (‘Late’ Duetz) was run in<br />

parallel to the flasks.<br />

This data indicates that although Duetz Microflasks can be used at an<br />

early screening stage to identify high producing clones the correlation<br />

with subsequent performance is improved after the cell lines are adapted<br />

to shaking growth conditions (ie after repeated subculture in shake<br />

flasks).<br />

Figure 1A shows that the top 8 cell lines identified in a ‘Late’ Duetz<br />

screen were the same as the top 8 lines identified in shake flasks. In<br />

contrast only 5 of the top 8 lines in the ‘Early’ Duetz screen were in the<br />

top 8 lines in shake flasks. Therefore the likelihood of discarding a high<br />

producing line is increased the earlier the Duetz screening is carried<br />

out.<br />

Similar results were obtained using a hydrolysate feed fed batch process<br />

in which the titre rank order and the response to feed in Duetz<br />

Microflasks was similar to that in shake flasks. Thus Figure 1B shows good<br />

correlation between the effect of the feed (% increase in titre) on cell<br />

lines in shake flasks and in ‘Late’ Duetz Microflasks and Figure 1C shows<br />

the correlation between the (titre) rank order in the two systems.<br />

Chemically defined medium process: A similar experiment was carried<br />

out with a different mAb producing CHO cell line in a chemically defined<br />

fed batch process. The standard (bioreactor and SF model) process<br />

involves a series of feeds during the fermentation. This multiple feed<br />

strategy was found to be impractical at the Microflask scale and so a<br />

compromise ‘single feed’ model was used in Duetz Microflasks. The<br />

correlation of rank order between ‘Early’ Duetz Microflasks and shake<br />

flasks was poor (R 2 = 0.34) but was significantly improved after Duetz<br />

Microflasks were inoculated with cells previously subcultured in shake<br />

flasks (R 2 = 0.73).<br />

Discussion: This data has demonstrated that, although there are some<br />

limitations around the adaptation of cell lines to shaking culture, Duetz<br />

Microflask screening can be used successfully to reduce the number of<br />

cell lines carried forward in the cell line selection process to shake flask<br />

or bioreactor screening.<br />

Although correlation with subsequent shake flasks is improved after<br />

repeated subculture (to allow complete adaptation to shaking) this data<br />

has also demonstrated that screening in Duetz Microflasks inoculated<br />

directly from static cultures or after minimal shake flask subculturing can<br />

be used to differentiate between cell lines that are high and low<br />

performers in conventional screening systems. We have used this system<br />

to reduce the number of cell lines carried forward to shake flask<br />

screening by approximately 70%.<br />

Page 36 of 181<br />

The simplicity of this system combined with the standard multiwell plate<br />

footprint facilitates automation and potentially enables large numbers of cell<br />

lines to be screened simultaneously and we are currently incorporating this<br />

system into our automated cell line generation process.<br />

P16<br />

Evaluation of an online biomass probe to monitor cell growth and cell<br />

death<br />

Angelo Perani 1* , Benjamin Gloria 1 , Dongmao Wang 1 , Anne-Sophie Buffier 2 ,<br />

Olivier Berteau 2 , Geoffrey Esteban 2 , Fiona E Smyth 1 , Andrew M Scott 1<br />

1 Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg,<br />

Victoria, 3084, Australia; 2 Fogale Nanotech, Nimes, 30900, France<br />

E-mail: angelo.perani@ludwig.edu.au<br />

BMC Proceedings 2011, 5(Suppl 8):P16<br />

Background: The estimation of cell density and cell viability of mammalian<br />

cell lines in cell culture has traditionally been performed using the exclusion<br />

dye trypan blue that stains “dead” cells when their cell membrane is<br />

damaged. In large scale cell cultures using bioreactors this estimation is<br />

performed off-line. The online biomass probe is based on the principle that<br />

under the influence of an electric field between two electrodes, ions in<br />

suspension migrate toward the electrodes. The cell plasma membrane is<br />

non-conductive so that the cells with intact plasma membranes are<br />

polarized and act as tiny capacitors and it has been shown that capacitance<br />

increases as the cell concentration does. The measurement is based on the<br />

linear relationship between the permittivity difference ε1- ε2 and the viable<br />

biomass concentration as it has been described by Ansorge et al. [1]. This<br />

study compares the data obtained using the biomass probe against the cell<br />

counts and viability determined by trypan blue exclusion with two GS-CHO<br />

cell line productions in bioreactors. Apoptosis determination measurements<br />

using rhodamine-123 and pan-caspase activation by flow cytometry will be<br />

compared to permittivity values.<br />

Material and methods: Cell Culture – Production: CHO-K1 SV cells were<br />

transfected with the vector coding for LICR C and LICR D antibodies using a<br />

Glutamine Synthetase expression vector (Lonza) and maintained in CD-CHO<br />

(Invitrogen) + 25mM of methionine sulfoximine (MSX) (Sigma). LICR C S1: 3L<br />

stirred-tank bioreactor (STR) (Applikon) in batch mode + temperature shift<br />

in CD-CHO + 32mM MSX (base 1); LICR C S2: 3L STR (Applikon) in batch<br />

mode + temperature shift in CD-CHO + 32mM MSX (base 2). LICR D S1: 3L<br />

STR (Applikon) in batch mode + 32mM MSX with addition of 4, 10 and<br />

20 mM NH4+. Biomass: Online monitoring was performed with an iBiomass<br />

Figure 1(abstract P16) Detection of apoptosis using a pan- caspase inhibitor labeled with FITC and rhodamine 123 during early stages of cell culture<br />

using flow cytometry in parallel with the online measurement of Δε with the online biomass probe.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

465 system (Fogale Nanotech). Viable Cell Concentration – Cell size: Viable<br />

Cell Concentration and cell size were estimated by off-line measurements<br />

using an automated trypan blue cell counter (Cedex, Roche). Apoptosis:<br />

Apoptosis related measurements were performed with a FACS Aria III<br />

(Becton Dickinson) using rhodamine-123 (Sigma) at 1mg/mL in buffered<br />

saline (PBS) for 10 6 cells or with 10 mM of CaspACE FITC-VAD-FMK<br />

(Promega) for 10 6 cells. FCS data was analysed with Gatelogic v3.08 (Inivai).<br />

Off-line measurement vs. on-line measurement: Correlation off-line and online<br />

measurements obtained by comparing the values of Xv.Dm4 to those of<br />

Δε (Xv: viable cell concentration estimated by Cedex in cells/mL; Dm:<br />

average diameter of cells estimated by Cedex in mm and Δε: cell<br />

permittivity measured by Fogale Biomass probe in pF/cm). Fluorescent<br />

Microscopy: 40μL of rhodamine 123 and CaspACE FITC-VAD-FMK stained<br />

samples were observed under fluorescent microscopy (FITC filter) with a 20x<br />

objective.<br />

Results: Permittivity vs. Cell Concentration: The comparison of the viable<br />

cell concentrations (Xv) profiles obtained with the Cedex and the<br />

permittivity profiles obtained with the biomass probe for the production<br />

of LICR C in bioreactors with different process controls show a clear<br />

correlation between the two measurements. The fitting of a linear<br />

regression curve gives a value of R 2 = 0.8777. (Data not shown).<br />

Diameter vs. permittivity related parameter: There is a direct correlation<br />

between the cell size (diameter) and viable cell concentration (Xv) measured<br />

by the Cedex and the permittivity measured with the biomass probe for the<br />

measurements obtained from LICR C bioreactor productions. The fitting of a<br />

linear regression curve gives a value of R 2 = 0.8798. (Data not shown).<br />

Apoptosis and permittivity: FACS analysis of a bioreactor run of LICR D cell<br />

lines under metabolic stress shows a different rhodamine-123 subpopulation<br />

distribution (forward scatter (FSC) vs. rhodamine 123 channel)<br />

less than 2h after first stress signal. There is no change in the caspase<br />

distribution (FSC vs. caspase). These results were confirmed with visual<br />

observation of the cells by fluorescence microscopy (Figure 1) using the<br />

same samples that were maintained in a FACS-fixing buffer: 16g of<br />

D-glucose (Sigma), 40% formaldehyde solution (Sigma) in 500 ml of 0.01M<br />

(1X) PBS pH7.2, stored at +4 0 C.<br />

Conclusion: The measurements obtained with the biomass probe<br />

demonstrated correlation between viable cell concentration and permittivity.<br />

These observations suggest that it is possible to use such a probe in the<br />

routine monitoring strategy for production of biopharmaceuticals using GS-<br />

CHO cell lines in bioreactors. Early detection of apoptosis in bioreactor<br />

cultures seems possible by using measurements obtained with the biomass<br />

probe. This type of measurements can be of critical value when developing<br />

processes that aim to minimise apoptosis. Future experiments need to be<br />

performed to correlate these observations with process parameter changes<br />

and mitochondrial modifications.<br />

Acknowledgement: The authors thank FOGALE Nanotech for the use of<br />

the iBiomass 465 unit and technical support.<br />

Reference<br />

1. Ansorge S, Esteban G, Schmid G: On-line monitoring of infected Sf-9<br />

insect cell cultures by scanning permittivity measurements and<br />

comparison with off-line biovolume measurements. Cytotechnology 2007,<br />

55:115-124.<br />

P17<br />

Impact on product quality of high productive GS-CHO cell lines<br />

Angelo Perani * , Benjamin Gloria, Dongmao Wang, Roger Murphy,<br />

Harjit Khangura Singh, Fiona E Smyth, Andrew M Scott<br />

Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg,<br />

Victoria, 3084, Australia<br />

E-mail: angelo.perani@ludwig.edu.au<br />

BMC Proceedings 2011, 5(Suppl 8):P17<br />

Background: The Clinical Program of the Ludwig Institute for Cancer<br />

Research (LICR) aims to translate basic laboratory discoveries into early<br />

phase clinical trials in cancer patients. A key component of the LICR<br />

approach has been to focus on the identification of antibodies selectively<br />

targeting antigens preferentially expressed in tumour tissue, and the<br />

molecular engineering of chimeric or humanised antibodies to these<br />

targets. The development of robust and high producing cell lines is<br />

crucial in the development of each antibody construct. The Cell Biology<br />

Group at the LICR Melbourne-Austin Branchisresponsibleforthe<br />

Page 37 of 181<br />

production of recombinant proteins including monoclonal antibodies<br />

from hybridomas or from industrially relevant mammalian cell lines (CHO,<br />

NS0, etc). The Cell Line Development activities are driven by the<br />

successful combination of industrial relevant cell lines transfected with<br />

cancer-relevant DNA targets using the Glutamine-Synthetase system (GS)<br />

from Lonza. Here we present two case-studies of high-productive cell<br />

lines in terms of product quality and biological activity of the protein<br />

produced with these cell lines.<br />

Material and methods: Cell line development / Production: CHO-K1 SV<br />

cells were transfected with the vector coding for monoclonal antibody<br />

LICR A within a GS expression vector (Lonza) and maintained in CD-CHO<br />

(Invitrogen) + 25μM methionine sulfoximine (MSX) (Sigma). LICR B (mu)<br />

murine hybridomas were generated against LICR B antigen and<br />

maintained in DMEM + 15% FBS (Invitrogen). Shake flask productivity<br />

screens were performed in E250 Erlenmeyer flasks with CD-CHO + 25μM<br />

MSX in batch mode for LICR A and LICR B and fed-batch for LICR B.<br />

Bioreactor productions: LICR A : 15L stirred-tanks bioreactors (STR)<br />

(Applikon) in batch mode + temperature shift in CD-CHO 32μM MSX;LICR<br />

B (mu): 15L STR in batch mode + temperature shift in DMEM (Invitrogen) +<br />

15% FBS and LICR B : 7L STR (Applikon) in fed-batch mode + temperature<br />

shift in CD-CHO + 32μM MSX. Purification: Supernatants were filtered and<br />

purified with Protein A eluted with 100mM glycine (pH 2.5), then adjusted<br />

to pH 8.0 with 1M Tris buffer then dialysed into PBS. ELISA assay: An antihuman<br />

IgG (g-chain specific) antibody for coating plates and an antihuman<br />

IgG (g-chain specific) alkaline phosphatase conjugate as secondary<br />

antibody. Purified antibody was used to set up a standard curve for<br />

calculating antibody concentration. Plates were read at 405 nm.<br />

SDS PAGE: Antibody (5μg) in a final volume of 20μl wasloadedunder<br />

both reducing and non-reducing conditions on NuPage, 4-12% Bis-Tris<br />

gels (Invitrogen) along with 20μl of molecular weight standards. Protein<br />

bands were detected with Coomassie blue staining. Biosensor analyses<br />

were conducted on a BIAcore 2000 instrument (GE). The epitope for the<br />

recombinant antibody was immobilized in a Ni-NTA chip and a blank<br />

control channel was for correction of refractive index effects. Sample at<br />

increasing concentrations were flowed over the chip surface and then<br />

washed out. The chip was regenerated with 0.1M EGTA between each<br />

analysis. FACS analysis was performed with a FACS Canto II cytometer<br />

(Becton Dickinson), Cell line expressing antigen A at 5x10 5 cells/sample;<br />

Negative control: isotype IgG (10μg /mL); Positive control: Purified LICR A<br />

(10μg/mL); Secondary antibody PE conjugated (1:1000) (Sigma); Data<br />

analysis using Gatelogic v 3.08 (Inivai).<br />

Results – discussion: LICR A: Production of the LICR A antibody<br />

increased from 265 mg/L in shake-flask production to 721 mg/L in 15L<br />

bioreactor (Results not shown). The FACS analysis of LICR A antibody with<br />

cells expressing the A antigen shows an increased binding when using<br />

purified LICR A antibody from the bioreactor production at 50 μg/mL<br />

(Mean Fluorescent Intensity (MFI) for PE from LICR A shake flask: 4591<br />

and MFI PE from LICR A bioreactor: 9396). Physicochemical analysis by<br />

SDS-PAGE of the product shows that the heavy and light chains (under<br />

reduced conditions) and the banding pattern (under non-reduced<br />

conditions) are typical for IgG. On SE-HPLC the main peak (94.7% for<br />

shake flask and 91.1% for bioreactor) is eluting at the expected size of an<br />

intact IgG.<br />

LICR B: Production of the LICR B antibody showed a continued increase<br />

from the production in bioreactor of the parental LICR B (mu): 109 mg/L<br />

to537mg/Linshake-flaskbatchmodeandto1252mg/Linshake-flask<br />

fed-batch mode (Results not shown). The highest productivity observed<br />

by ELISA was observed with the supernatant from the 7L bioreactor in<br />

fed-batch mode: 2244 mg/L. The BIAcore comparative analysis of the<br />

material purified from the LICR B (mu) bioreactor and LICR B bioreactor<br />

fed-batch shows an increased binding (decreased K d):KdLICRB(mu)<br />

bioreactor: 0.41 nM; K d LICR B bioreactor fed-batch: 1.13 pM (Figure 1).<br />

Physicochemical analysis by SDS-PAGE of the product shows that the<br />

heavy and light chains (under reducedconditions)andthebanding<br />

pattern (under non-reduced conditions) are typical for IgG. On SE-HPLC<br />

the main peak [96.8% for LICR B (mu) and 95.9% for LICR B] is eluting as<br />

an intact IgG (Results not shown).<br />

Conclusion: We have successfully constructed two GS-CHO cell lines<br />

producing functional intact antibodies against cancer-related epitopes.<br />

The product concentration was substantially increased for both cell lines<br />

without having a negative effect of product quality. Analyses of complex<br />

glycans present on LICR A and LICR B antibodies are to be performed.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P17) Binding to cells expressing the LICR A antigen analysed by FACS for purified LICR A antibody and Binding to LICR B antigencoated<br />

chips analysed by BIAcore for purified LICR B antibody.<br />

P18<br />

Anti-diabetes effect of water containing hydrogen molecule and Pt<br />

nanoparticles<br />

Sanetaka Shirahata 1,2* , Takeki Hamasaki 1 , Keisuke Haramaki 2 ,<br />

Takuro Nakamura 1 , Masumi Abe 1 , Hanxu Yan 2 , Tomoya Kinjo 2 ,<br />

Noboru Nakamichi 3 , Shigeru Kabayama 3 , Kiichiro Teruya 1,2<br />

1 Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu<br />

University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan; 2 Division of<br />

Life Engineering, Graduate School of Systems Life Sciences, Kyushu<br />

University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan; 3 Nihon<br />

Trim Co. Ltd., 1-8-34 Oyodonaka, Kita-ku, Osaka 531-0076, Japan<br />

E-mail: sirahata@grt.kyushu-u.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):P18<br />

Background: Electrochemically reduced water (ERW) contains a lot of<br />

hydrogen molecule (H 2) and scavenges reactive oxygen species (ROS) to<br />

protect DNA from oxidative damage [1]. ERW also contains small amounts of<br />

Pt nanoparticles (NPs) and elongates the lifespan of C. elegans [2]. Pt NPs are<br />

newly recognized multi-functional ROS scavengers [3]. ERW exhibits antidiabetes<br />

effects in vitro and in vivo [4-6][7]. We proposed mineral<br />

nanoparticle active hydrogen reduced water hypothesis to explain the<br />

Page 38 of 181<br />

activation mechanism of H 2 to hydrogen atom (H)[4]. Recently, H 2 has been<br />

reported to scavenge ROS and suppress a variety of oxidative stress-related<br />

diseases [8], however, the action mechanism of H 2 has not been clarified<br />

thoroughly. Here, we examined anti-diabetes effects of H2 and Pt NPs.<br />

Materials and methods: Pt NPs of 2-3 nm sizes were synthesized from<br />

H 2PtCl 6 by the citrate reduction method. L6 rat myoblast cells (1.2 x 10 5<br />

cells) were inoculated into a 35 mm culture dish and a day later, the cells<br />

were treated with or without 25mM N-acetylcystein in the presence of BES-<br />

H2O2, a H 2O 2-specific detection reagent in DMEM for 2 h. After washing the<br />

cells, molecular hydrogen treatment was performed in a dark condition by<br />

cultivating cells in a fresh DMEM medium in a mixed gas incubator under an<br />

atmosphere of 75%N 2/20%O 2/5%CO 2 or 75%(H 2 and N 2 mixed gas)/20%O 2/<br />

5%CO 2 for 1.5 h, followed by flowcytometric analysis. In this condition,<br />

culture medium contained maximum 0.4-0.5 ppm of dissolved hydrogen.<br />

Glucose uptake of differentiated myotube L6 cells was examined after<br />

treating the cells with 3 H-2-deoxyglucose for 10 min. Gene expression of<br />

catalase (CAT), glutathione peroxidase (GPx) and hemoxoigenase (HO-1) was<br />

examined using RT-PCR method. Three weeks old type 2 diabetes model<br />

mice (KK-A y )werefedH 2 and/or Pt Nps-containing water ad lib for 6 weeks.<br />

Results: H 2 stimulated glucose uptake into L6 cells. Pt NPs catalyzed the<br />

activation of H 2 to hydrogen atom (H) to scavenge DPPH radical in vitro.<br />

The combined use of molecular hydrogen and Pt NPs resulted in


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

extremely stimulated glucose uptake into L6 cells, suggesting that H<br />

produced from H 2 by catalyst action of Pt NPs regulated glucose uptake<br />

signal transduction. As oppose to the paper by Ohsawa et al.[8], H 2 of 25<br />

to 75% concentration in the mixed gas significantly scavenged<br />

intracellular H 2O 2 in rat fibroblast L6 cells (Figure 1) and induced the<br />

gene expression of antioxidative enzymes such as CAT, GPx and HO-1 via<br />

activation of Nrf2 (Figure 2). H 2, Pt NPs and their combination<br />

significantly suppressed the levels of fasting blood glucose and improved<br />

Figure 1(abstract P18) The scavenging effect of hydrogen molecule on intracellular hydrogen peroxide in rat myotube L6 cells. ***, p


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

the impaired sugar tolerance abilities of obese insulin-resistant type 2<br />

diabetic KK-A y mice.<br />

Conclusion: H 2, Pt NPs, and their combined use resulted in activation of<br />

glucose uptake signal transduction pathways and stimulation of glucose<br />

uptake into L6 myotubes. In the groups of H2, Pt NPs and their combined<br />

use groups, blood sugar levels and impaired sugar tolerance of type 2<br />

diabetes model mouse (KK-A y ) were significantly improved, suggesting that<br />

H 2, Pt NPs and H are redox regulation factors in animal cells.<br />

References<br />

1. Shirahata S, Kabayama S, Nakano M, Miura T, Kusumoto K, Gotoh M,<br />

Hayashi H, Otsubo K, Morisawa S, Katakura Y: Electrolyzed-reduced water<br />

scavenges active oxygen species and protects DNA from oxidative<br />

damage. Biophys Biochem Res Commun 1997, 234:269-274.<br />

2. Yan H, Tian H, Kinjo T, Hamasaki T, Tomimatsu K, Nakamichi N, Teruya K,<br />

Kabayama S, Shirahata S: Extension of the lifespan of Caenorhabditis<br />

elegans by the use of electrolyzed reduced water. Biosci Biotech Biochem<br />

2010, 74:2011-2015.<br />

3. Hamasaki T, Kashiwagi T, Imada T, Nakamichi N, Aramaki S, Toh K,<br />

Morisawa S, Shimakoshi H, Hisaeda Y, Shirahata S: Kinetic analysis of<br />

superoxide anion radical-scavenging and hydroxyl radical-scavenging<br />

activities of platinum nanoparticles. Langmuir 2008, 24:7354-7364.<br />

4. Shirahata S, Hamasaki H, Teruya K: Advanced research on the health<br />

benefit of reduced water. Trends Food Sci Tech 2011, DOI 10.1016/j.<br />

tifs.2011.10.009.<br />

5. Li Y–P, Nishimura T, Teruya K, Maki T, Komatsu T, Hamasaki T, Kashiwagi T,<br />

Kabayama S, Shim S–Y, Katakura Y, Osada K, Kawahara T, Otsubo K,<br />

Morisawa S, Ishii Y, Gadek Z, Shirahata S: Protective mechanism of<br />

reduced water against alloxan-induced pancreatic b-cell damage:<br />

Scavenging effect against reactive oxygen species. Cytotechnology 2002,<br />

40:139-149.<br />

6. Li Y–P, Hamasaki T, Nakamichi N, Kashiwagi T, Komatsu T, Ye J, Teruya K,<br />

Abe M, Yan H, Kinjo T, Kabayama S, Kawamura M, Shirahata S: Suppressive<br />

effects of electrolyzed reduced water on alloxan-induced apoptosis and<br />

type 1 diabetes mellitus. Cytotechnology 2010, DOI 10.1007/s10616-010-<br />

9317-6.<br />

7. Kim M–J, Kim H–K: Anti-diabetic effects of electrolyzed reduced water in<br />

streptozotocin-induced and genetic diabetic mice. Life Sciences 2006,<br />

79:2288-2292.<br />

8. Ohsawa I, Ishikawa M, Takahashi K, Watanabe M, Nishimaki K, Yamagata K,<br />

Katsura K, Katayama Y, Asoh S, Ohta S: Hydrogen acts as a therapeutic<br />

antioxidant by selectively reducing cytotoxic oxygen radials. Nature Med<br />

2007, 13:688-694.<br />

P19<br />

Metabolic enhancers: a new paradigm in cell culture media<br />

optimization<br />

Abi M Abitorabi 1* , Christopher Wilcox 2<br />

1 2<br />

GIRUS Life Sciences, Inc., Sunnyvale, CA, 94085, USA; Sheffield BioScience,<br />

Beloit, WI, 53511, USA<br />

E-mail: abi@girusinc.com<br />

BMC Proceedings 2011, 5(Suppl 8):P19<br />

Our primary focus is enabling and accelerating biological drug<br />

manufacture through the development of cost-effective technologies that<br />

Page 40 of 181<br />

facilitate rapid bioprocess development and improve manufacturing<br />

“bang-for-the-buck”. Using data mining and cell-based screens we have<br />

designed the Regocel small molecule yield enhancers for a number of<br />

mammalian cell platforms used in biomanufacturing such as CHO and NS0<br />

cells. Chemically defined molecules were screened against several<br />

parameters: growth, viability and most importantly protein production.<br />

Formulations were developed that are defined, animal-free, non-nutritional<br />

and compatible with different media, nutritional supplements, culture<br />

formats and scales. Because they target ubiquitous cellular pathways such<br />

as apoptosis, cell cycle and protein synthesis, these formulations provide<br />

consistent results with a variety of clones and can shorten bioprocess<br />

times and cost. Our results with Regocel supplements demonstrated<br />

increased protein production in a variety of commercial media (Figure 1),<br />

increased yields of different proteins such as antibodies and mammalian<br />

target of rapamycin (mTOR), immediate yield enhancements (from passage<br />

1), and persistence of yield enhancements over many passages with no<br />

permanent alterations to cells, as determined by return of productivity to<br />

control levels upon Regocel supplement removal from the medium.<br />

These new small molecule formulations provide a new means of<br />

improving cell culture outcome independently of mammalian cell clone,<br />

cell culture medium and process.<br />

P20<br />

Neuroprotective effects of 3,5-di-o-caffeoylquinic acid in vitro and<br />

in vivo<br />

Junkyu Han 1,2 , Hiroko Isoda 1,2*<br />

1 Graduate School of Life and Environmental Sciences, University of Tsukuba.<br />

Tsukuba, Ibaraki 305-8572, Japan; 2 Alliance for Research on North Africa<br />

(ARENA), University of Tsukuba. Tsukuba, Ibaraki 305-8572, Japan<br />

E-mail: isoda.hiroko.ga@u.tsukuba.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):P20<br />

Background: Caffeoylquinic acid (CQA) derivatives are natural functional<br />

compounds isolated from a variety of plants and possess a broad range of<br />

pharmacological properties, including antioxidant, hepatoprotectant,<br />

antibacterial, antihistaminic, anticancer, and other biological effects [1].<br />

Recently, it has been demonstrated that CQA derivatives possess<br />

neuroprotective effects in Ab-induced PC12 cell toxicity and in<br />

tetrahydropapaveroline (THP)-induced C6 glioma cell death [2]. One of the<br />

animal models that is used to study AD and aging is the senescenceaccelerated<br />

mouse (SAM). The SAM model was developed in 1981, which<br />

originally consisted of nine major senescence-accelerated-prone mice<br />

(SAMP) substrains and three major senescence-accelerated-resistant mice<br />

(SAMR) substrains, each of which exhibits the characteristic disorders.<br />

Methodology: As in vitro experiment, the human neuroblastoma clonal<br />

SH-SY5Y cell were maintained at 37°C under 5% CO 2 /95%air.Asin vivo<br />

experiment, the CQA-treated mice were orally administered with 3,5-di-O-<br />

CQA mixed with drinking water (6.7 mg/kg · day) for 1 month using oral<br />

administration tube and syringe. Proteomics analysis, real-time PCR,<br />

measurement of intracellular ATP content, Moris water maze were carried<br />

out to investigate the neuroprotective effect of CQA.<br />

Results: 3,5-di-O-CQA had neuroprotective effect on Ab 1–42 treated cells.<br />

The mRNA expression of glycolytic enzyme (phosphoglycerate kinase-1;<br />

PGK1) and intracellular ATP level were increased in CQA treated SH-SY5Y<br />

Figure 1(abstract P19) Antibody production by Chinese hamster ovary (CHO) cells increase with the Regocel small molecules (RS-CHO) in different<br />

commercial media (M1-M6).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P20) Effect of CQA on the spatial learning and memory of SAMP8 mice in MWM.<br />

cells. We also found that CQA administration induced the improvement of<br />

spatial learning and memory on SAMP8 mice, and the overexpression of<br />

PGK1 mRNA.<br />

Conclusion: CQA has a neuroprotective effect on Ab1–42 treated SH-SY5Y<br />

cells. The mRNA expression of glycolytic enzyme (PGK1) and the intracellular<br />

ATP level were increased in CQA-treated SH-SY5Y cells. We also found that<br />

CQA administration induced the improvement of spatial learning and<br />

memory on SAMP8 mice, and the overexpression of PGK1 mRNA level.<br />

These findings suggest that CQA has a neuroprotective effect through the<br />

induction of PGK1 expression and ATP production activation.<br />

References<br />

1. Basnet P, Matsushige K, Hase K, Kadota S, Namba T: Four di-O-caffeoyl<br />

quinic acid derivatives from propolis. Potent hepatoprotective activity in<br />

experimental liver injury models. Biol Pharm Bull 1996, 19:1479-1484.<br />

2. Soh Y, Kim JA, Sohn NW, Lee KR, Kim SY: Protective effects of quinic acid<br />

derivatives on tetrahydropapaveroline induced cell death in C6 glioma<br />

cells. Biol Pharm Bull 2003, 26:803-807.<br />

P21<br />

Improvement of insulin resistance by Cyanidin 3-glucoside, anthocyanin<br />

from black beans through the up-regulation of GLUT4 gene expression<br />

Tetsuya Inaguma 1 , Junkyu Han 1,2 , Hiroko Isoda 1,2*<br />

1 Graduate School of Life and Environmental Sciences University of Tsukuba,<br />

Tsukuba, Japan; 2 Alliance for Research on North Africa (ARENA) University of<br />

Tsukuba, Tsukuba, Japan<br />

E-mail: isoda.hiroko.ga@u.tsukuba.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):P21<br />

Introduction: Black beans have been suggested to have a protective effect<br />

against obesity. Cyanidin 3-glucoside (Cy-3-G) belongs to the flavonoid class<br />

of molecules and is a member of the anthocyanin family that is present in<br />

black beans. Some studies indicated that Cy-3-G from black beans may be<br />

beneficial for the improvement of obesity and type Ⅱ diabetes. It has been<br />

reported that Cyanidin 3-glucoside ameliorates insulin sensitivity due by<br />

down-regulating the retinol binding protein 4 expression in diabetic mice [1].<br />

Accumulation of neutral lipids, triglyceride (TG) in particular, is highly related<br />

to the development of insulin resistance and its consequences, such as type<br />

Ⅱ diabetes. Lipid droplet size regulation is central in the regulation of<br />

metabolism and in adipocytokines secretion such as TNF-a and adiponectin<br />

[2,3]. However, little is currently known about how Cy-3-G influences<br />

adipocyte differentiation and insulin resistance in 3T3-L1 adipocytes. The<br />

purpose of the present study was to determine the effects of Cy-3-G on<br />

GLUT4 gene expression to improve insulin resistance in 3T3-L1 adipocytes.<br />

Materials and methods:<br />

Cell culture and treatment: Adipocytes, 3T3-L1 cells (Riken Cell Bank,<br />

Ibaraki, Japan), were maintained in Dulbecco’s modified Eagle’s medium<br />

(DMEM) (Sigma Chemical Co, St. Louis, MO, USA) supplemented with 10%<br />

Page 41 of 181<br />

fetal bovine serum (FBS) (Biowest, Miami, FL, USA, Japan) and 1% 5,000<br />

units/ml penicillin, 5,000 μg/mL streptomycin (PS) (Lonza Walkersville,<br />

MD,USA) and incubated in 37 °C in 5% CO 2. For adipocyte differentiation,<br />

3T3-L1 cells were cultured at 3.0×10 5 cells/well in 6-well plates. Cells were<br />

maintained until full confluence, which is often around 48h, prior to<br />

treatment, . And then, cells were treated with differentiation medium<br />

containing Dexamethazone (DEX) Solution, 3-Isobuthyl-1-Methylxanthine<br />

(IBMX) Solution, Insulin Solution, and Cy-3-G. DEX Solution, IBMX Solution,<br />

and Insulin Solution were purchased from Cayman Chemicals (Ann. Arbor,<br />

MI, USA). Cy-3-G derived from black soybean was purchased from Fujicco<br />

Co., Ltd. (Kobe, Japan). After 72 h of induction, medium was changed to<br />

DMEM containing insulin and Cy-3-G was added every 2 days. After 4<br />

days of incubation from the initiation of differentiation, bioassays were<br />

performed.<br />

RNA extraction and real-time PCR: Total RNA was isolated from the 3T3-<br />

L1 cells using ISOGEN (Nippon Gene, Tokyo, Japan) according to the<br />

manufacturer’s instructions. The integrity of the RNA extracted from all<br />

samples was verified and quantified using NANO DROP 2000 (Agilent<br />

Technologies, CA). Total RNA was used for the single strand cDNA<br />

synthesis with a cDNA synthesis kit, SuperScript® III Reverse Transcriptase.<br />

Gene expression levels were analyzed by quantitative real-time PCR, using<br />

the Applied Biosystems 7500 FAST INSTURMENT (Applied Biosystems,<br />

Foster City, CA, U.S.A.). The oligonucleotide primers of mouse were<br />

obtained from Applied Biosystems (Foster City, CA, U.S.A.). The cDNA was<br />

denatured at 95 °C for 10 min, followed by 40 cycles of PCR (95 °C, 15 sec;<br />

60 °C, 60 sec).<br />

Results and discussion: In Cy-3-G-treated cells, the number of droplets<br />

increased, while the lipid droplet size decreased. Cy-3-G significantly<br />

promoted the mRNA expressions of peroxisome proliferrator-activated<br />

receptor-g (PPARg) [4], CCAAT/enhancer binding protein a (C/EBPa) [4],<br />

and glucose transport 4 (GLUT4) [Table 1] in differentiated 3T3-L1 cells.<br />

PPARg and C/EBPa mainly control adipocyte differentiation. GLUT4 takes in<br />

glucose on cell membrane. According to the results, Cy-3-G promotes<br />

adipocyte differentiation and uptake of glucose in a dose-dependent<br />

manner. In the present study, the concentrations of Cy-3-G treated were<br />

20 μM, and 100 μM. Cy-3-G at 20 μM, and 100 μM significantly decreased<br />

TNF-a concentration and ROS production, and increased the adiponectin<br />

concentration in differentiated 3T3-L1 cell in a dose-dependent manner<br />

[4]. These effects suggest that Cy-3-G would contribute to the<br />

improvement of insulin resistance and its consequences. However, when<br />

we utilize black beans as food, the doses of Cy-3-G in black beans would<br />

be lower than the concentrations that we used in this study. Therefore, the<br />

study on the effects of Cy-3-G at lower concentrations, such as 1 μM, 5 μM<br />

and 10 μM, on adipocyte differentiation and adipocytokines secretion in<br />

differentiated 3T3-L1cells will be needed. Also, the anti-fat effects of Cy-3-<br />

G in vivo using diabetic model mice have not yet been widely reported.<br />

From our study, Cy-3-G derived from black bean as a food factor is<br />

expected to prevent life style-related disease.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P21) Effect of Cy-3-G on the gene<br />

expression of GLUT4 in 3T3-L1 adipocytes<br />

Sample Relative GLUT4 gene expression level<br />

(% of Control)<br />

Control 100 ± 1.5<br />

Cy-3-G 20 µM 208.2 ± 1.4 *<br />

Cy-3-G 100<br />

226.57 ± 15.2<br />

µM<br />

*<br />

Cy-3-G significantly induced the gene expression of GLUT4 in a dosedependent<br />

manner. *p< 0.05 (vs Control).<br />

Conclusion: Cy-3-G has the potential to improve insulin resistance and its<br />

consequences in 3T3-L1 adipocytes through the up-regulation of GLUT4<br />

gene expression. Additional study on insulin resistance using 3T3-L1and<br />

anti-fat effect using diabetic model mice will be needed to verify these<br />

results in vivo.<br />

Acknowledgement: This research was partially supported by JST-JICA’s<br />

Science and Technology Research Partnership for Sustainable<br />

Development (SATREPS).<br />

References<br />

1. Sasaki R, Nishimura N, Hoshino H, Isa Y, Kadowaki M, Ichi T, Tanaka A,<br />

Nishiumi S, Fukuda I, Ashida H, Horio F, Tsuda T: Cyanidin 3-glucoside<br />

ameliorates hyperglycemia and insulin sensitivity due to downregulation<br />

of retinol binding protein 4 expression in diabetic mice. Biochem Pharm<br />

2007, 74:1619-1627.<br />

2. Okuno A, Tamemoto H, Tobe K, Ueki K, Mori Y, Iwamoto K, Umesono K,<br />

Akanuma Y, Fujiwara T, Horikoshi H, Yazaki Y, Kadowaki T: Troglitazone<br />

increases the number of small adipocytes without the change of white<br />

adipose tissue mass in obese Zucker rats. J Clin Invest 1998,<br />

101:1354-1361.<br />

3. Kadowaki T: Insights into insulin resistance and type 2 diabetes from<br />

knockout mouse models. J Clin Invest 2000, 106:459-465.<br />

4. Han J, Inaguma T, Isoda H: Cyanidin 3-glucoside anthocyanin from black<br />

beans has potential to protect insulin resistance on 3T3-L1 adipocytes<br />

by inhibiting TNF-a release. Br J Nutr , (in press).<br />

Page 42 of 181<br />

P22<br />

Engineering CHO cell growth by stable manipulation of miRNA expression<br />

Noëlia Sanchez * , Nga Lao, Clair Gallagher, Martin Clynes, Niall Barron<br />

National Institute for Cellular Biotechnology, Dublin City University, Dublin 9,<br />

Ireland<br />

BMC Proceedings 2011, 5(Suppl 8):P22<br />

Background: MiRNAs are small non-coding RNAs involved in many<br />

biological functions such as cell proliferation and apoptosis (1), cell cycle (2),<br />

homeostasis (3) and cell metabolism (4). They are highly conserved between<br />

species. They are capable of regulating hundreds of genes in a posttranscriptional<br />

manner by translation repression and/or mRNA degradation<br />

(5). These characteristics make miRNAs attractive tools for CHO cell<br />

engineering as multiple genes may be targeted simultaneously and possibly<br />

entire biological pathways can be manipulated. After temperature-shifting<br />

CHO cell culture from 37°C to 31°C, several miRNAs were found differentially<br />

regulated of which miR-7 was found to be down-regulated. Our laboratory<br />

has previously demonstrated that transient overexpression of miR-7 using<br />

mimics of mature endogenous miR-7, leads to a significant decrease in cell<br />

growth (6). On the other hand, transient inhibition using inhibitors of<br />

mature endogenous miR-7 provoked increased cell growth, though to a<br />

lesser extent. As this antisense technology is transient, another strategy was<br />

chosen to study the impact of stable miR-7 inhibition on CHO phenotypes.<br />

“Sponge” technology is an effective tool to stably sequester endogenous<br />

miRNAs (7) by introducing a decoy target reporter gene. This approach can<br />

be used to understand post-transcriptional regulation in CHO cells and to<br />

identify cellular pathways related to cell growth, cell viability and cell<br />

productivity - key phenotypes for cell bioprocess improvement.<br />

Materials and methods: Binding sites for either miR-7 (sponge miR-7) or a<br />

non-specific control sequence (sponge NC) were inserted into pCMV-deGFP<br />

(Professor.Sharp’s laboratory). SEAP-secreting CHO-K1 cells were transfected<br />

with 5µg of these constructs in a 6-well plate. Selection pressure was<br />

applied on day 1 after transfection by addition of hygromycin at 350µg/ml.<br />

Cells were transferred to a T-75 flask. From the mixed population, cells with<br />

high and medium GFP positivity were selected for single cell sorting using<br />

FACS flow cytometer in 96-well plates. Expanded clones were transferred to<br />

24-well plates in adherent and suspension culture. GFP positivity and cell<br />

growth were monitored by Guava EasyCyte flow cytometer.<br />

Figure 1(abstract P22) CHO-K1 SEAP cell growth in sponge NC and sponge miR-7 expressing cell clones at day 3 of cell culture.P-value=0.0024<br />

in student t-test.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Results: Design of sponge NC/miR-7: Four tandemly repeated miR-7<br />

binding sites were cloned downstream of a destabilised enhanced GFP to<br />

increase efficacy of endogenous miRNA sequestration. The negative<br />

control consisted of the same cassette with non-specific sequences to<br />

replace miR-7 binding sites. The deGFP, a modified enhanced GFP,<br />

consists of a fusion of the ornithine decarboxylase degradation domain<br />

from mouse (MODC) and the C-terminal of an enhanced variant of GFP<br />

(eGFP) conferring a shorter half-life than eGFP (2h). The change of deGFP<br />

fluorescence induced by the binding of endogenous miR-7 to the sponge<br />

can be correlated to the change in active miRNA levels. To avoid RNAitype<br />

cleavage by Argonaute 2 and degradation, the sponges were<br />

designed with a bulge region (position 9-11).<br />

Impact of sponge miR-7 on CHO phenotypes in a mixed population:<br />

After transfection of the sponge miR-7 and sponge NC in SEAP-secreting<br />

CHO-K1 cells, GFP positivity and cell growth were monitored in both mixed<br />

populations and single clones. In mixed populations, sponge miR-7<br />

transfected cells were found to have reduced GFP positivity compared to<br />

control pools (sponge miR-7: 34.9% and control 56.1%) and significantly<br />

improved cell density at day 3 (1.6x10 6 cells/ml versus 1.16x10 6 cells/ml<br />

respectively) and at day 4 of culture (3.4x10 6 cells/ml and 2.9x10 6 cells/ml,<br />

respectively).<br />

Impact of sponge miR-7 on CHO phenotypes in single clones: In single<br />

clones, the average of GFP positivity was lower in sponge miR-7 expressing<br />

clones than in control clones (49.6% compared to 30.9%). The average cell<br />

density for 23 clones was significantly higher in sponge miR-7 expressing<br />

clones than in the control expressing clones (1.3x10 6 cells/ml and<br />

1.01x10 6 cells/ml). Moreover, 12 clones from the sponge miR-7 expressing<br />

clones achieved more than 1.4x10 6 cells/ml whereas only one clone from<br />

the control expressing cells could reached this density. (Figure 1).<br />

Conclusions: In this study, we showed that miRNAs may be used as tools<br />

to improve CHO phenotypes, in this case cell density, and also as potential<br />

tools for understanding of CHO cellular pathway regulation. The application<br />

of miRNA expression manipulation in large-scale culture could be a novel<br />

technology to increase significantly the performance of biopharmaceutical<br />

production processes.<br />

Acknowledgements: This work was supported by NICB, DCU and<br />

Science Foundation Ireland.<br />

References<br />

1. Cheng AM, Byrom MW, Shelton J, Ford LP: Antisense inhibition of human<br />

miRNAs and indications for an involvement of miRNA in cell growth and<br />

apoptosis. Nucleic Acids Res 2005, 33(4):1290-7.<br />

2. Lal A, Navarro F, Maher CA, Maliszewski LE, Yan N, O’Day E, et al: miR-24<br />

inhibits cell proliferation by targeting E2F2, MYC, and other cell-cycle<br />

genes via binding to “seedless” 3 ‘ UTR MicroRNA recognition elements.<br />

Mol Cell 2009, 35(5):610-25.<br />

3. Li X, Cassidy JJ, Reinke CA, Fischboeck S, Carthew RW: A MicroRNA imparts<br />

robustness against environmental fluctuation during development. Cell<br />

2009, 137(2):273-82.<br />

4. Gao P, Tchernyshyov I, Chang T, Lee Y, Kita K, Ochi T, et al: c-myc<br />

suppression of miR-23a/b enhances mitochondrial glutaminase<br />

expression and glutamine metabolism. Nature 2009, 458(7239):762-U100.<br />

5. Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, et al:<br />

Microarray analysis shows that some microRNAs downregulate large<br />

numbers of target mRNAs. Nature 2005, 433(7027):769-73.<br />

6. Barron N, Kumar N, Sanchez N, Doolan P, Clarke C, Meleady P, et al:<br />

Engineering CHO cell growth and recombinant protein productivity by<br />

overexpression of miR-7. J Biotechnol 2011, 151(2):204-11.<br />

7. Ebert MS, Neilson JR, Sharp PA: MicroRNA sponges: Competitive<br />

inhibitors of small RNAs in mammalian cells. Nature Methods 2007,<br />

4(9):721-6.<br />

P23<br />

Expression of recombinant human coagulation factors VII (rFVII) and IX<br />

(rFIX) in various cell types, glycosylation analysis, and pharmacokinetic<br />

comparison<br />

Ernst Böhm 1* , Michael Dockal 1 , Michael Graninger 1 , Meinhard Hasslacher 1 ,<br />

Martin Kaliwoda 1 , Christian Konetschny 1 , Artur Mitterer 1 , Eva-Maria Muchitsch 2 ,<br />

Manfred Reiter 1 , Friedrich Scheiflinger 2<br />

1 2<br />

Baxter BioScience, Orth/Donau, A-2304, Austria; Baxter BioScience, Vienna,<br />

A-1220, Austria<br />

BMC Proceedings 2011, 5(Suppl 8):P23<br />

Page 43 of 181<br />

Introduction: Clearance mechanisms for rFVII (or the active enzyme rFVIIa)<br />

and rFIX are influenced by post-translational modifications, especially<br />

N-glycosylation. This should be considered when choosing a recombinant<br />

expression system in view of the varying ability of frequently used cell lines<br />

to perform modifications similar to human proteins.<br />

Differences in the pharmacokinetic properties of recombinant FVIIa versus<br />

plasma-derived (pd)FVII or desialylated rFVIIa are known for human FVII(a).<br />

Asialo rFVIIa clears quickest, whereas pdFVII, having a higher degree of<br />

sialylation, is cleared to a lesser extent [1]. In the case of FIX, the degrees<br />

ofserinephosphorylationandtyrosinesulfationintheactivationpeptide<br />

have been postulated to influence pharmacokinetic behavior, especially<br />

in vivo recovery [2].<br />

We chose CHO, BHK and HEK293 cells for expression of rFVII to compare<br />

post-translational protein modifications, and HEK293-derived cell lines to<br />

generate highly phosphorylated and sulfated rFIX for in vivo studies. rFIX<br />

from the same clone and production run was purified using two different<br />

down-stream processes: The first to enrich high phosphorylated and<br />

sulfated protein, the second to purify total rFIX at high yield. These HEK293derived<br />

rFIX isoforms were compared with CHOrFIX and pdFIX in a<br />

pharmacokinetic study in FIX knock-out mice.<br />

Materials and methods:<br />

Proteins: pdFVII and pdFIX were from HTI, recombinant, CHO-derived FIX<br />

was from Wyeth.<br />

Cell culture: BHK (BHK-21; ATCC#CCL-10) and HEK293 cells (ATCC#1531)<br />

were from American Type Culture Collection, CHO DXB11 from University<br />

of Columbia. All were cultivated in DMEM/Ham’s F12 medium containing<br />

fetal bovine serum (FBS). BHK and HEK293 cell-derived rFVII producer<br />

clones were selected by antibiotic resistance. CHO DXB11-derived<br />

producer clones were generated by methotrexate gene co-amplification.<br />

rFVII was produced in vitamin K-containing medium without FBS.<br />

HEK293 cells producing human FIX were selected by antibiotic resistance.<br />

Clones were adapted to serum-free suspension culture in Excell293 medium<br />

containing vitamin K, and cultivated in repeated batch mode fermentation<br />

runs.<br />

Purification: rFVII was purified using Q-Sepharose FF for capture, and<br />

a tandem step containing a cellufine sulfate column connected to<br />

Q-Sepharose FF.<br />

HEK293-derived rFIX from the same clone from one fermentation run was<br />

purified from culture supernatants with two different purification schemas:<br />

for high yield, rFIX was loaded in the presence of EDTA on Q-Sepharose FF,<br />

the column was washed with EDTA at high salt concentration, FIX was<br />

eluted at low salt CaCl2. For enrichment of the highly phosphorylated and<br />

sulfated protein fraction, rFIX was loaded in the presence of EDTA on<br />

fractogel TMAE, washed with EDTA at high salt concentration, and eluted<br />

with a salt gradient in the presence of CaCl 2.<br />

Analytics: Purified FVII forms were analyzed using reversed phase HPLC,<br />

LC-MS for intact protein or peptide mapping after trypsin digestion to<br />

confirm sequences and post-translational modifications, and anion<br />

exchange HPLC for monosaccharide composition, sialic acid quantification,<br />

and comparison of N-glycans.<br />

Degrees of FIX-phosphorylation/sulfation were monitored by LC-MS. Nglycosylation<br />

was characterized by oligosaccharide mapping (HPLC<br />

method).<br />

In vivo pharmacokinetic study: FIX preparations were administered to<br />

FIX-knock-out mice intravenpusly at a nominal dose of 75 U/kg. Citrate<br />

plasmas were obtained by heart puncture. FIX levels in plasmas were<br />

determined by antigen enzyme-linked immunoassay and by activated<br />

partial thromboplastine time (APTT) clotting assay.<br />

Results and conclusions: Identical protein structures and minor differences<br />

in post-translational modifications between rFVII from BHK, CHO, HEK293<br />

cells, and pdFVII were found, except for N-glycosylation. Protein activities of<br />

rFVII determined by antigen and activity assays were similar for each cell<br />

type. Most N-glycans of pdFVII are bi- and triantennary, non-fucosylated<br />

structures with almost complete sialylation [3]. In contrast, all rFVII forms<br />

were sialylated only in part. Approximately 50% N-glycans of BHKrFVII were<br />

composed of a core-fucosylated biantennary complex-type with two<br />

terminal sialic acids. Another 20% contained terminal GalNAc, a sugar<br />

moiety with a high affinity for the asialoglycoprotein receptor involved in<br />

the in vivo clearance of glycoproteins from circulation. GalNAc were not<br />

found on N-glycans on CHO-derived rFVII. Most oligosaccharides on<br />

CHOrFVII were core fucosylated, biantennary structures carrying two sialic<br />

acid residues. HEK293rFVII showed major differences from all other rFVII


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P23) AUCs of pdFIX, CHOrFIX, HEK293rFIX, and high-phospho/sulfo HEK293rFIX in a FIX-knock-out<br />

mouse pharmacokinetic study. Only relative AUC values are shown. Statistically significant different ratios to CHOrFIX<br />

(at the 5 % level) are marked with an asterisk*. The ratio of HEK293rFIX high phospho/sulfo to HEK293- total rFIX was<br />

also statistically significantly different for both antigen and activity<br />

Relative value of AUC: antigen Relative value of AUC: clotting activity<br />

pdFIX 1.5* 1.8*<br />

CHOrFIX 1 1<br />

HEK293rFIX high phospho/sulfo 0.7* 0.7*<br />

HEK293rFIX 0.4* 0.4*<br />

forms, and, unexpectedly, from pdFVII. Most oligosaccharide structures were<br />

not comparable to those published for pdFVII [3], or found on rFVII from<br />

the other cell lines. A high content of fucosylation and terminal GalNAc,<br />

and a lower degree of terminal sialic acids were measured, and tri- or tetraantennary<br />

structures were not found. Some oligosaccharides were of a<br />

hybrid type with high-mannose structures. These N-glycan structures do not<br />

favor the use of HEK293 cells as a production platform for human<br />

biotherapeutics, when protein recovery and half-life in the circulation are of<br />

importance, and influenced by N-glycosylation.<br />

Pharmacokinetic differences between pdFIX and CHO-derived rFIX were<br />

as expected from the literature; HEK293rFIX materials (total rFIX and<br />

high phospho/sulfo rFIX) were both inferior to CHO-derived rFIX in terms of<br />

area under the curve (AUC) (Table 1) and in vivo recovery. Similar terminal<br />

half-lives and mean residence times, but different in vivo recoveries between<br />

rFIX and pdFIX preparations were confirmed by statistical analysis. High<br />

phospho/sulfo rFIX from HEK293 had a larger AUC and in vivo recovery than<br />

total rFIX from the same HEK293-derived clone and fermentation run,<br />

indicating an influence of these modifications on pharmacokinetics (Figure 1).<br />

Oligosaccharide mapping showed high glycosylation heterogeneity<br />

for HEK293 and pdFIX, and more defined peak-groups representing mono- to<br />

tetrasialylated glycans for CHO-derived material. Sialic acid content of Nglycans<br />

was 25% lower for HEK293- than for CHOrFIX. Glycosylation and<br />

phosphorylation/sulfation degrees contributed to pharmacokinetics of FIX<br />

preparations. If glycosylation was similar in case of the two HEK293rFIX<br />

preparations, the effects of phosphorylation and sulfation became evident.<br />

In conclusion, these data showed that HEK293 cells were not adequate for<br />

rFIX or rFVII production due to improper N-glycosylation compared with the<br />

material from other cell lines, or from human plasma. Consequently, an<br />

Page 44 of 181<br />

advantage of this human cell line for the production of “more-human-like”<br />

biotherapeutics could not be observed.<br />

References<br />

1. Appa RS, Theill C, Hansen L, Møss J, Behrens C, Nicolaisen EM, Klausen NK,<br />

Christensen MS: Investigating clearance mechanisms for recombinant<br />

activated factor VII in a perfused liver model. Thromb Haemost 2010,<br />

104(2):243-251.<br />

2. Ewenstein BM, Joist JH, Shapiro AD, Hofstra TC, Leissinger CA, Seremetis SV,<br />

Broder M, Mueller-Velten G, Schwartz BA, Mononine Comparison Study<br />

Group: Pharmacokinetic analysis of plasma-derived and recombinant F<br />

IX concentrates in previously treated patients with moderate or severe<br />

hemophilia B. Transfusion 2002, 42(2):190-197.<br />

3. Fenaille F, Groseil C, Ramon C, Riandé S, Siret L, Chtourou S, Bihoreau N:<br />

Mass spectrometric characterization of N- and O-glycans of plasmaderived<br />

coagulation factor VII. Glycoconj J 2008, 25(9):827-842.<br />

P24<br />

Complex medium supplements optimized for the reduction of<br />

animal-derived components in vaccine production media<br />

James F Babcock * , Karen A Benedict, Amanda L Perlman<br />

Sheffield Center for Cell Culture Technology, Sheffield Bio-Science, A Kerry<br />

Group Business, Ithaca, NY USA<br />

E-mail: james.babcock@kerry.com<br />

BMC Proceedings 2011, 5(Suppl 8):P24<br />

Introduction: A series of cell-line specific complex media supplements<br />

have been developed for enhancing performance of various vaccine<br />

Figure1(abstract P23) Pharmacokinetic comparison of pdFIX, CHOrFIX, HEK293rFIX, and high-phospho/sulfo HEK293rFIX in FIX-knock-out mice. Plasma<br />

concentrations of FIX after administration are shown as mean values ± standard deviations with n = 10 mice for each timepoint. Dose adjustment was<br />

done by normalizing to start material concentrations.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

production systems. Created using in-house media optimization methods,<br />

these supplements are manufactured using innovative process technology<br />

which allows for the formulation of complex and/or chemically<br />

defined animal-component free media additives into a single homogenized<br />

functional supplement. These supplements have been optimized<br />

for individual cell lines, and have proven to be suitable for use in a range<br />

of basal media. Data are presented which demonstrate the effectiveness<br />

of these optimized supplements for application as performance<br />

enhancers, and as a vehicle for serum-reduction in CEF, MDCK and BHK<br />

culture media. While these particular supplements do contain some<br />

undefined components, preliminary research indicates that this same<br />

technology may be applied to chemically-defined, multi-component<br />

supplements.<br />

Materials and methods: CEF and MDCK cells were maintained in T-75<br />

flasks with a working volume of 15 ml. CEF cells were grown in DMEM +<br />

5% Tryptose Phosphate Broth (TPB) + 5% FBS. MDCK cells were grown in<br />

DMEM + 10% FBS.<br />

To generate growth curves for both CEF and MDCK, triplicate T-25 flasks<br />

were seeded at 2.0 x10 5 cells/ml with a working volume of 7.5 ml. Cells were<br />

incubated at 37°C in 5% CO 2 . One set of triplicates was set up for each day<br />

cells were to be counted. On days 3-6, one set of triplicates was trypsinized<br />

and re-suspended in fresh medium for counting.<br />

BHK cultures were grown in 125 ml shake-flasks containing a final medium<br />

volume of 35 ml. The basal medium consisted of DMEM plus 10% FBS.<br />

Triplicate cultures were seeded at 2.0 x10 5 cells/ml, and incubated at 37°C<br />

in 5% CO 2 at 130 rpm for 12 days. Hydrolysate supplementation was<br />

achieved via the use of filter-sterilized 100 g/l stock solutions prepared in<br />

the basal medium.<br />

On days 3-7, 1.0 ml of the culture supernatants were removed for assessing<br />

cell counts and viability. All cells were counted using a Nova BioProfile Flex<br />

automated cell counter.<br />

Results: In a medium for Chicken Embryo Fibroblast (CEF) cells, incorporation<br />

of non-animal components into a complex supplement allows for<br />

reduction in the final concentration of serum in the basal medium<br />

formulation. This results in a significant reduction in animal-derived products<br />

as compared to the traditional CEF medium formulation containing 5%TBP +<br />

5% FBS. The non-animal supplement out-performs the conventional<br />

medium with respect to cell growth when the serum level is reduced to 1%<br />

(Figure 1).<br />

As with the CEF cells, various hydrolysates were screened to determine<br />

the most compatible with MDCK cells. Sheff-VAX is a hydrolysate-based<br />

animal component free supplement which can be employed to reduce<br />

FBS concentration from 10% to 3% or less. When Sheff-VAX is used in<br />

conjunction with 3% FBS, higher cell densities are achieved than with the<br />

control medium containing 10% FBS.<br />

In BHK-21 culture, supplementation with Sheff-VAX or Sheff-VAX Plus ACF<br />

allows for significantly improved cell counts while simultaneously<br />

Page 45 of 181<br />

reducing the serum requirement. The control media for both cell lines<br />

contain 10% FBS, and both cell lines reached similar maximum cell<br />

densities. However, the magnitude of improvement achieved with the<br />

animal component free supplements was significantly greater with the<br />

BHK-21 cells as compared to the MDCK cells.<br />

Summary: The data presented illustrate how complex animal component<br />

free supplements may be employed to reduce or eliminate serum from<br />

many traditional vaccine production media, while providing equal or<br />

better performance with respect to cell growth. Partial or complete serum<br />

elimination was achieved in media used to cultivate CEF, MDCK and BHK-<br />

21 cell lines. These supplements have also proven to facilitate the<br />

transition from anchorage dependent to suspension culture.<br />

P25<br />

Application of complex and chemically-defined medium supplements<br />

toward cell line specific performance enhancement of<br />

biopharmaceutical production systems<br />

James F Babcock * , Karen A Benedict, Amanda L Perlman<br />

Sheffield Center for Cell Culture Technology, Sheffield Bio-Science, A Kerry<br />

Group Business, Ithaca, NY USA<br />

E-mail: james.babcock@kerry.com<br />

BMC Proceedings 2011, 5(Suppl 8):P25<br />

Introduction: A series of cell-line specific complex media supplements have<br />

been developed for enhancing performance of various biopharmaceutical<br />

production systems. Created using in-house media optimization methods,<br />

these supplements are manufactured using innovative, proprietary process<br />

technology which allows for the combination of complex and/or chemically<br />

defined animal-component free media additives into a single homogeneous<br />

functional supplement. These supplements have been optimized for<br />

individual cell lines, and have proven to be suitable for use in a range of<br />

basal media. Data are presented which demonstrate the effectiveness of<br />

these optimized supplements as performance enhancers for application in<br />

CHO and SP2/0 batch culture, and as feed supplements in fed-batch<br />

systems. Preliminary data will also be presented on a parallel series of strictly<br />

chemically-defined supplements, similarly optimized for specific cell lines.<br />

Materials and methods: CHO data were collected using a transfected<br />

CHO-K1 line (ATCC #CCL-61), adapted to serum-free suspension culture, and<br />

engineered to constitutively express secreted embryonic alkaline<br />

phosphatase (SEAP) by means of a modified human cytomegalovirus<br />

(hCMV) promoter.<br />

Hybridoma data were collected using a murine hybridoma suspension cell<br />

line (ATCC # CRL-1753). This line was derived from primary spleen cell<br />

cultures of animals immunized with purified human immunoglobulin.<br />

These spleen cells were then fused with Sp2/0-Ag14 myeloma cells to<br />

create the hybridoma.<br />

Figure 1(abstract P24) Growth performance of an animal component free medium supplement in Chicken Embryo Fibroblast (CEF) adherent cell culture.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Triplicate 125 ml shake-flasks contained a final medium volume of 35 ml.<br />

Duplicate spinner flasks had a working volume of 150 ml. The basal<br />

medium consisted of 100% chemically defined medium (CDM). Cultures<br />

were incubated at 37°C in 5% CO2. Medium supplement stock solutions<br />

were prepared at 100 g/l in the basal medium and sterilized through a<br />

2.0 µm filter.<br />

At appropriate points during each experiment, 1.0 ml of the culture<br />

supernatants were removed for assessing cell counts and viability. Cells<br />

were counted using a Nova BioProfile Flex automated analyzer. On the<br />

final day of each run, 500 µl of the culture supernatants were removed<br />

for SEAP or IgG analysis. Levels of SEAP in the supernatants were<br />

measured using anion-exchange HPLC on a Waters Model 2695 HPLC<br />

Figure 1(abstract P25) Performance comparison of defined and undefined medium supplements in CHO batch culture.<br />

Page 46 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

separations module equipped with a dual-wavelength absorbance<br />

detector. IgG was quantified using a Protein G affinity column.<br />

Results: Using in-house media optimization methods, both an undefined<br />

and a chemically-defined supplement for CHO cells were developed from<br />

a variety of individual components. Themostfavorabledosageforeach<br />

supplement was determined through a series of dose-response<br />

experiments. While both supplements improved cell culture performance,<br />

the undefined supplement achieved a significantly higher peak cell<br />

density than the chemically defined supplement (Figure 1).<br />

Both SEAP titer and specific productivity (Qp) were improved when the<br />

supplements were employed (Figure 1). As reflected in the growth data,<br />

the undefined provided a significantly greater benefit. While the titer<br />

increase for the chemically-defined supplement was substantial, there<br />

was little increase in the Qp. The significant Qp increase seen with the<br />

undefined supplement undoubtedly accounts for the higher SEAP titer<br />

obtained, and may be a function of un-characterized elements within the<br />

hydrolysate based supplement.<br />

Summary: Medium optimization is an integral part of biopharmaceutical<br />

process development. There is an on-going debate within the industry as<br />

to the various advantages and disadvantages of both defined and<br />

undefined media and media components. The choice of which type of<br />

system to employ is often motivated by risk mitigation with respect to<br />

consistency of performance, as weighed against the underlying goal of<br />

achieving the highest possible product titers for any given system. Defined<br />

and undefined supplementation solutions may not be mutually exclusive.<br />

P26<br />

A comparison of performance enhancing synergy among<br />

ultrafiltered yeast extracts and recombinant human serum<br />

albumininCHO-K1cells<br />

James F Babcock * , Karen A Benedict, Amanda L Perlman<br />

Sheffield Center for Cell Culture Technology, Sheffield Bio-Science, A Kerry<br />

Group Business, Ithaca, NY USA<br />

E-mail: james.babcock@kerry.com<br />

BMC Proceedings 2011, 5(Suppl 8):P26<br />

Introduction: We have previously demonstrated a synergistic reaction<br />

between a wheat hydrolysate and recombinant human serum albumin used<br />

to supplement a chemically defined growth medium for SP2/0 hybridoma<br />

cells. The data presented here illustrate the synergystic performance<br />

enhancing effect obtained when ultrafiltered yeast extract and recombinant<br />

human serum albumin are co-supplemented in CHO cell media. Each<br />

combination has its own distinctive effect on the growth and productivity of<br />

transfected cells. Cell viability, cell proliferation and target protein<br />

production all may be improved, yet these effects are not necessarily<br />

observed concurrently in a given system.<br />

Materials and methods: Data were collected using a transfected CHO-K1<br />

line, adapted to serum-free suspension culture, and engineered to<br />

constitutively express secreted embryonic alkaline phosphatase (SEAP) by<br />

means of a modified human cytomegalovirus (hCMV) promoter.<br />

Figure 1(abstract P26) Growth Curves and SEAP Titers for rHSA, HyPep YE and UltraPep YE Supplemented Batch Cell Cultures.<br />

Page 47 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Cultures were grown in 125 ml shake-flasks containing a final medium<br />

volume of 35 ml. The basal medium consisted of 100% chemically defined<br />

medium (CDM) supplemented with 1 mg/ml G-418. Triplicate flasks were<br />

seeded at 4.0 x10 5 cells/ml, and incubated at 37°C in 5% CO2 at 130 rpm<br />

for 12 days. Medium supplement stock solutions were prepared at 100 g/l<br />

in the basal medium and sterilized through a 2.0 µm filter.<br />

At days 5, 7, 8, 9 and 12, 1.0 ml of the culture supernatants were<br />

removed for assessing cell counts and viability. Cells were counted using<br />

a Nova BioProfile Flex automated analyzer. At Day 12, 1.0 ml of the<br />

culture supernatants were removed for SEAP analysis. Levels of SEAP in<br />

the supernatants were measured using anion-exchange HPLC.<br />

Results: Maximum cell density increased with respect to the Medium<br />

Control when cultures were dosed with rHSA at 1 g/l, but not when<br />

supplemented with HyPep YE at 1 g/l. When both supplements were<br />

used together, an even greater increase in cell density was observed. The<br />

synergystic effect was also seen with rHSA and UltraPep YE. However, the<br />

UltraPep YE/rHSA combination out-performed the HyPep YE/rHSA with<br />

respect to maximum cell density (Figure 1). All cultures were assayed for<br />

total SEAP production on Day 12. When dosed at 1g/l, all of the<br />

supplements (HyPep YE, UltraPep YE and rHSA) yielded higher titers than<br />

the Medium Control. The greatest increases were seen when HyPep YE or<br />

UltraPep YE were used in conjunction with rHSA (Figure 1).<br />

Summary: The data presented here illustrate the performance-enhancing<br />

synergy that may be realized by supplementing various cell culture<br />

media with a combination of yeast extract and recombinant human<br />

Page 48 of 181<br />

serum albumin. When the two supplements are used together, cell<br />

culture performance results exceed those achieved when using each<br />

supplement individually. Overall performance was further improved by<br />

varying the individual dosages of yeast extract and recombinant human<br />

serum albumin. In four separate basal media, cell response to cosupplementation<br />

for each of the yeast extract/recombinant albumin<br />

combinations tested was shown to be both medium and dosage<br />

dependent. The optimized combination provided significant overall<br />

performance improvement in all media tested.<br />

P27<br />

Targeted metabolomics for bioprocessing<br />

Denise Sonntag * , Francesca M Scandurra, Torben Friedrich, Michael Urban,<br />

Klaus M Weinberger<br />

BIOCRATES Life Sciences AG, Innsbruck, Austria<br />

E-mail: denise.sonntag@biocrates.com<br />

BMC Proceedings 2011, 5(Suppl 8):P27<br />

Background: Bioprocesses like the cell-based production of biologicals, i.e.<br />

mainly recombinant proteins and monoclonal antibodies, require optimal<br />

culture conditions to obtain a high yield of quality products. The<br />

performance of a bioreactor highly depends on the cell characteristics as well<br />

as on the composition of the cell culture medium and the process<br />

conditions. As the metabolic activity of the cells is very high during<br />

Figure 1(abstract P27) Comparison of the metabolite spectrum of commercially available cell culture media supplements derived from biological sources.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

fermentation, the external and internal metabolite compositions vary<br />

tremendously throughout the process. The quantification of a wide range of<br />

metabolic substrates and products is a prerequisite to understand and<br />

optimize the underlying cell-based activities. Furthermore, metabolite<br />

quantification reveals the composition of biologically derived cell culture<br />

supplements, thus serving as a tool to monitor supplement quality or<br />

providing the base for the formulation of a chemically defined medium<br />

supplement.<br />

Materials and methods: Targeted metabolomics was carried out using<br />

a mass spectrometry-based platform. Analyses were performed with<br />

commercially available KIT plates that allow the simultaneous quantification<br />

of 180 metabolites [1]. The fully automated assay is based on PITC<br />

(phenylisothiocyanate)-derivatization in the presence of internal standards<br />

followed by FIA-MS/MS (for acylcarnitines, lipids, hexoses) as well as LC-MS/<br />

MS (amino acids and biogenic amines) using a AB SCIEX 4000 QTrap mass<br />

spectrometer in the multiple reaction monitoring detection mode with<br />

electrospray ionization. On the same instrument, a validated HPLC-MS/MS<br />

quantification method is used for the analysis of energy metabolism<br />

intermediates. The concentrations of individual fatty acids are determined as<br />

their corresponding methyl ester derivatives (FAME’s) using GC-MS on an<br />

Agilent 7890 GC/5795 MSD instrument after derivatization. Vitamins were<br />

determined using LC-ESI-MS/MS technique after their pre-separation into two<br />

fractions (water- and fat-soluble vitamins).<br />

Results: The metabolomics approach to bioprocess monitoring made it<br />

possible to determine the concentration of a multitude of analytes from<br />

several metabolite classes in a sample- and time-efficient way by using small<br />

sample volumes and a multi-assay strategy. The analyte portfolio went<br />

beyond routinely determined culture media components, as it covered<br />

proteinogenic and non-proteinogenic amino acids, e.g. ornithine and<br />

citrulline, and intermediates of the energy metabolism, e.g. lactate, hexoses,<br />

succinate, as well as acylcarnitines, biogenic amines, glycerophospholipids,<br />

sphingomyelins, fatty acids, and vitamins (Figure 1).<br />

Conclusions: Targeted metabolomics comprises a rapid and comprehensive<br />

method to determine the composition of cell culture supplements of<br />

biological origin.<br />

The method is also well suited to rapidly characterize fermentational<br />

processes metabolically by monitoring changes in medium composition and<br />

cellular metabolite pools. The received quantitative data can be directly<br />

related to growth, vitality, and productivity of the cell.<br />

Reference<br />

1. [http://www.freepatentsonline.com/20070004044.html].<br />

P28<br />

Toolbox approach for fast generation of stable CHO production cell<br />

lines from different hosts<br />

Susanne Seitz * , Henning von Horsten, Thomas Rose, Volker Sandig,<br />

Karsten Winkler<br />

ProBioGen AG, Berlin, 13086, Germany<br />

E-mail: Susanne.Seitz@probiogen.de<br />

BMC Proceedings 2011, 5(Suppl 8):P28<br />

Figure 1(abstract P28) Overview of PBG Cell Line Development.<br />

Page 49 of 181<br />

Background: Protein production in CHO cell lines is a highly dynamic<br />

process, where the yield limiting step can shift among transcription,<br />

translation, and secretion under different cell growth stages and culture<br />

conditions. In addition, the choice of CHO cell line has been shown to affect<br />

target protein quality and quantity.<br />

Further advancements in titer and specific productivity require elimination<br />

of cellular/process bottlenecks along the generation of stable production<br />

cell lines including vector design, clone selection, genetic engineering,<br />

and media and feed optimization.<br />

To overcome these limitations, we developed a toolbox for all process<br />

phases from transfection to production. This toolbox is based on a<br />

chemically defined media platform and includes pre-adapted CHO-K1 or<br />

CHO-DG44 cell lines, optimized vectors for single and multi-chain proteins<br />

as well as fine-tuned protocols. In addition, it enables the production of<br />

antibodies with specialized glycan structures (GlymaxX® [1]) as well as clone<br />

engineering with regard to the expression and secretion machinery (CDC42).<br />

Materials and methods: ProBioGen‘s toolbox expression vectors<br />

encoding an Fc-protein or therapeutic glycoprotein were generated using<br />

gene-optimized sequences and optimized signal peptides.<br />

To create stable cell lines, expression vectors were transfected into CHO-<br />

DG44 and CHO-K1 cells by microporation. Two serum-free selection<br />

strategies were applied using both MTX and puromycin: selection in<br />

semi-solid medium followed by automated clone picking and deposition<br />

into 96 wells using the ClonePix FL (Genetix), and combined selection<br />

and cloning in 96-well plates. For a first expression analysis monoclonal<br />

primary clones were subjected to 96-well plate assay. Clones showing<br />

best performance in 96-well were expanded and taken into batch and<br />

fed-batch for productivity assessment. Best performing primary clones<br />

were subjected to a second cloning step by limiting dilution and<br />

analyzed in 96-well. Following clone scale-up highest ranking sub-clones<br />

clones were assessed in shake flask using fed-batch. Prior to master cell<br />

banking candidate lead clones were tested for fed-batch bioreactor<br />

performance in a Cellferm-pro® (DASGIP AG). Protein concentrations were<br />

determined using either an ELISA method, the Gyrolab SIA Ligand<br />

Binding Assay or an HPLC method.<br />

Protein secreted from stable cell lines was purified either by protein A<br />

affinity chromatography (Fc-protein) or Ion Exchange Chromatography<br />

using weak anion exchangers (glycoprotein) and evaluated by Size-<br />

Exclusion HPLC (SEC), Western blot and SDS-PAGE.<br />

To determine the stability of clones a continuous passage of roughly 80<br />

population doublings with analyzing protein batch titers as well as mRNA<br />

levels and gene copy numbers (StepOnePlus RT-PCR System) at defined<br />

time points was performed. Cells were cultivated without selection<br />

pressure.<br />

Results and conclusion: Here we describe an innovative parallel<br />

platform cell line development for CHO-K1 and CHO-DG44 (Figure 1)<br />

exemplified by two completely different recombinant proteins, an Fcprotein<br />

and a therapeutic glycoprotein, with the glycoprotein showing<br />

differences in glycosylation pattern in the two starter cell lines.<br />

Our toolbox approach enables a fast and highly reproducible generation<br />

of high producer clones stably expressing the transgene for more than 80


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P28) Growth and protein production data of CHO producer clones in different assay formats<br />

Fc-protein Glycoprotein<br />

Assay format Cell line Max VCD [viable cells/mL] Max Titer [mg/L] Max VCD [viable cells/mL] Max Titer [mg/L]<br />

96-well plate assay K1 nd a<br />

50 nd a<br />

268<br />

(5 days) DG44 nd a<br />

176 nd a<br />

607<br />

Batch K1 2.3E+07 610 1.1E+07 1772<br />

(5 or 7 days) DG44 8.6E+06 980 6.4E+06 2200<br />

Fed batch K1 3.0E+07 2500 1.9E+07 8100<br />

(12-17 days) DG44 1.7E+07 4500 3.0E+07 13200<br />

a not determined.<br />

generations without selection pressure and resulting in upstream yields of<br />

4.5-13 g/L (Table 1).<br />

Acknowledgement: We would like to thank the PBG teams for their hard<br />

and excellent work.<br />

Reference<br />

1. von Horsten HH, Ogorek C, Blanchard V, Demmler C, Giese C, Winkler K,<br />

Kaup M, Berger M, Jordan I, Sandig V: Production of non-fucosylated<br />

antibodies by co-expression of heterologous GDP-6-deoxy-D-lyxo-4hexulose<br />

reductase. Glycobiology 2010, 20:1607-1618.<br />

P29<br />

Growth characterization of CHO DP-12 cell lines with different high<br />

passage histories<br />

Christoph Heinrich 1* , Timo Wolf 1 , Christina Kropp 1 , Stefan Northoff 2 ,<br />

Thomas Noll 1<br />

1<br />

Institute of Cell Culture Technology, Bielefeld University, Bielefeld, Germany;<br />

2<br />

TeutoCell AG, Bielefeld, Germany<br />

E-mail: che@zellkult.techfak.uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P29<br />

Introduction: For industrial pharmaceutical protein production fast<br />

growing, high producing and robust cell lines are required. To select pHshift<br />

permissive and faster growing sub-populations, the CHO DP-12 cell line<br />

was serially subcultured for more than four hundred days in shaker flasks.<br />

Initial adaptation to growth in suspension was carried out in chemically<br />

defined medium without hypoxanthine and thymidine (HT), while the final<br />

medium used for long term cultivation contains HT. Cell samples were<br />

cryopreserved at four different time points after 21, 95, 165 and 420 days.<br />

Cultivations of these four sub-populations (SP) in shaker flasks and<br />

bioreactors revealed considerable differences in specific growth rates<br />

and product formation as well as in the metabolism of glucose, lactate and<br />

several amino acids. For the elucidation of the intracelluar mechanism<br />

behind these alteration in growth characteristics and metabolism additional<br />

probes were analyzed using proteomic and metabolomic approaches [1].<br />

Material and methods: In this study the CHO DP-12 clone#1934 (ATCC<br />

CRL-12445) was used as reference organism. It co-expresses the variable<br />

light and heavy chains of the murine 6G4.2.5 monoclonal antibody (ATCC-<br />

HB-11722) which inhibits binding of interleukin 8 to human neutrophile.<br />

CHO DP-12 cells were cultivated in CD-ACF medium TC 42 (TeutoCell AG)<br />

and PowerCHO-2 (LONZA AG) for the first steps of suspension adaptation.<br />

200 nM methotrexate was present at any time. Precultures and parallel<br />

cultivations were carried out in 125 mL and 250 mL polycarbonate<br />

Erlenmeyer flasks (Corning Life Sciences). Incubator conditions were set to<br />

37°C, 5% CO 2 and relative humidity of 80%. A shaker revolution of 185 rpm<br />

or 125 rpm with an orbital movement of 2” was chosen. For bioreactor<br />

cultivations four parallel vessels (Applikon) controlled by CellfermPro 2.3<br />

software (DASGIP AG) were used. Cultivation parameters were set to 37°C,<br />

40% DO and pH 7.1.<br />

Cell concentration and viability were determined with a CEDEX system<br />

(Innovatis-Roche AG). The anti IL-8 antibody was quantified using Protein A<br />

HPLC.<br />

A MACSQuant® Analyzer (Miltenyi Biotec GmbH) was used for the<br />

measurements of intracellular IgG-product pools. Intracellular detection of<br />

the antibodies required permeabilization of the cell membrane with<br />

detergents. IgG light and heavy chains were stained with fluorochrome-<br />

Page 50 of 181<br />

conjugated antibodies that bind to Fc and kappa chains of IgGs within<br />

fixed CHO cells (all solutions from Miltenyi Biotec GmbH).<br />

Results: The initial adaptation to growth in suspension of the CHO DP-12<br />

cells led to considerable changes in average cell diameter and specific<br />

growth rate. Within the first 100 days of serial subculturing the cell<br />

diameter dropped from a maximum of about 17 µm to 12 µm as the<br />

lowest value. In the same period the cell specific growth rate increased<br />

from initially 0.2 d -1 up to values greater than 1.0 d -1 . After further 320<br />

days cells had an average diameter about 14 ± 0.74 µm and a mean<br />

specific growth rate of 0.82 ± 0.12 d -1 .<br />

The growth characterizations of the four sub-populations SP21, SP95, SP165<br />

and SP420 were carried out in several parallel controlled and uncontrolled<br />

batchcultivations.Inadditiontotheinfluenceoftheinsulinlikegrowth<br />

factor 1 (IGF) the presence of hypoxanthine and thymidine (HT) was<br />

investigated as well. Most important characteristics for growth and<br />

productivity determined in this experimental setup are presented in Figure 1.<br />

Though the sub-populations SP95, SP165 and SP420 seemed to possess<br />

comparable growth rates in shake flask cultivations, they differed<br />

remarkably in their maximum cell density when HT-mix was present. For<br />

SP21 and SP420, maximum cell densities of about 1.1·10 7 cells/mL and<br />

2.1·10 7 cells/mL, respectively, were determined. These results indicate an<br />

influence of the HT-mix on the maximum reachable cell densities.<br />

Furthermore, an increased passage number seemed to cause decreasing<br />

growth performance in the controlled bioreactor system compared to the<br />

results from the shake flask cultivations with HT containing medium. This<br />

fact could be associated to the adaptation of subcultivated cells to pHshifts<br />

that occur during cultivation under uncontrolled conditions.<br />

Additionally, the observed differences between the four sub-populations<br />

in terms of the metabolism of glucose, lactate and several amino acids<br />

might play a role in this context. In bioreactor cultivations a two-fold<br />

higher lactate formation was observed for SP420 as compared to SP21,<br />

for instance.<br />

The highest specific production rate of 10.5 pg/(cell·day) was obtained for<br />

the sub-population SP95 resulting in a final antibody titer of 334 mg/L.<br />

Considering long-term cultivation, cell specific productivity increased<br />

during first passages and was lost with ongoing subcultivation. Further<br />

cytometry analysis regarding intracellular productivity of examined CHO<br />

DP-12 cells revealed that serial subculturing resulted in accumulation of a<br />

subclone expressing only the light chain of the IL-8 antibody.<br />

Conclusions: Serial subculturing over an extended period led to the<br />

selection of faster growing and pH-shift permissive cells, thus resulting in<br />

higher viable cell densities, especially in uncontrolled shake flask<br />

cultivations. Furthermore, cells or rather sub-populations with distinct<br />

metabolic characteristics were enriched along the subculturing process.<br />

This indicated that a targeted experimental approach could be used e.g.<br />

to specifically select cells adapted to low glutamine concentrations and<br />

therefore, a reduced consumption rate. On the other hand, the observed<br />

loss of productivity shows that the selection pressure given by 200 nM<br />

methotrexate and deprivation of hypoxanthine and thymidine could not<br />

prevent an increase of sub-populations expressing no or only incomplete<br />

(non-native, deficient) product. This problem could be caused by the<br />

vector design maybe used for the CHO DP-12 cells, which resulted in a<br />

non associated integration of the dihydrofolate reductase (dhfr) and anti-<br />

IL 8 sequences. Hence, it might be interesting to monitor the intracellular<br />

expression during clone selection or even seed-train development by<br />

flow cytometry.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P29) Overview of growth performance (A) and product formation (B) of the four sup-populations in controlled and uncontrolled<br />

culture systems with varying media supplementation.<br />

Acknowledgements: The authors would like to thank Miltenyi Biotec<br />

GmbH for providing the MACSQuant® analyzer and consumables.<br />

Reference<br />

1. Beckmann T, Thüte T, Heinrich C, Büntemeyer H, Noll T: Proteomic and<br />

metabolomic characterization of CHO DP-12 cells with different high<br />

passages histories. Proc 22nd ESACT Meeting Vienna 2011.<br />

P30<br />

Utilization of multifrequency permittivity measurements in addition to<br />

biomass monitoring<br />

Christoph Heinrich * , Tim Beckmann * , Heino Büntemeyer, Thomas Noll<br />

Institute of Cell Culture Technology, Bielefeld University, 33615 Bielefeld,<br />

Germany<br />

E-mail: che@zellkult.techfak.uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P30<br />

Introduction: In recent years measurement of permittivity signal has been<br />

increasingly used for online biomass monitoring of cell cultures. Breweries<br />

use it as an established method for fermenter inoculation and bioprocess<br />

controlforinstance[1].Inthecaseofanimalcellculturesthecorrelation<br />

between permittivity and viable cell densities determined offline varies<br />

along cultivation time. Hence, several authors have used the permittivity<br />

signal as an indirect method for measuring oxygen and glutamine<br />

consumption as well as intracellular nucleotide phosphate concentrations<br />

[2,3]. The latest generation of biomass monitoring devices allows parallel<br />

measurement of permittivity at a range of frequencies leading to an<br />

improvement in the correlation between biomass and permittivity and<br />

providing a tool to further explore other aspects related to the physiological<br />

state of the cells.<br />

Material and methods: In this study 10 different cell lines, among them<br />

industrial cell lines as well as cell lines distributed by ATCC, were cultivated.<br />

Cell type specific chemically defined and animal component-free media<br />

(TeutoCell AG) were used. Batch, fed-batch and perfusion bioreactor<br />

cultivations were carried out in controlled benchtop vessels (Sartorius AG or<br />

Applikon Biotechnology). The i-Biomass 465 sensor (FOGALE nanotech) was<br />

used for online multifrequency permittivity monitoring. In addition the<br />

permittivity signal was used to implement a fully automated cell bleed to<br />

maintain a constant viable cell density in a perfusion process. Furthermore, a<br />

fed-batch feed strategy was introduced to keep the substrate concentration<br />

at a certain level. Cell density and viability were determined using a CEDEX<br />

system (Innovatis-Roche AG). Glucose and lactate were measured with an<br />

YSI 2700 Biochemistry Analyzer (YSI Life Sciences). Amino acids were<br />

quantified using an in-house developed HPLC method.<br />

Results: The FOGALE i-Biomass 465 sensor was used to monitor the<br />

viable cell density of different human, CHO and hybridoma cell cultures<br />

Page 51 of 181<br />

online. A good correlation of the permittivity signal and the offline<br />

measured viable cell density for the growth phase was verified (R > 0.99),<br />

but pH-shifts and increased cell aggregation had a negative impact on<br />

the correlation. The linear factor to calculate the viable cell density from<br />

the online permittivity signal varied between 4.5·10 5 cells/(pF/cm) and<br />

12.0 cells/(pF/cm). A clear relation between cell type (CHO, human or<br />

hybridoma) and the linear factor could not be established from the<br />

available data.<br />

Subsequently, the online biomass monitoring system was used to carry out<br />

a 1 L spin-filter perfusion process with constant viable cell density at a<br />

predefined setpoint. The application of a permittivity closed-loop controlled<br />

cell bleed resulted in a steady concentration of 10 7 viable cells/mL during<br />

perfusion, at a dilution rate of 1.0 d -1 . As soon as this threshold was reached,<br />

the cell bleed was automatically started and controlled based on the online<br />

signal of the i-Biomass 465 sensor.<br />

In addition to the correlation with viable cell density, a linear relationship<br />

(R 2 > 0.96) between the online i-Biomass 465 signal and the concentrations<br />

of numerous components, e.g. glucose, lactate, asparagine, glutamine,<br />

tyrosine, threonine, methionine, lysine, phenylalanine, serine, leucine and<br />

isoleucine, was found during the exponential growth phase of CHO-K1 and<br />

CHO DP-12 cultivations. The results indicated that the number of<br />

correlating substrates depended on the used cell line (CHO, human or<br />

hybridoma) and the process strategy (constant pH or pH-shift). Since, the<br />

established substrate correlations were more robust against process<br />

variations, they were investigated as a basis for a closed-loop feeding<br />

strategy in fed-batch cultivations. Compared to a pre-defined feeding<br />

schedule or to intermitted feeding this would have the advantage of<br />

avoiding nutrient limitations and substrate accumulation that might occur<br />

due to unexpected high or low cell growth. Also, feeding would be<br />

independent of human surveillance. The successful application of a<br />

completely automated permittivity-controlled feeding strategy was proved<br />

in two fed-batch runs with CHO DP-12 (ATCC CRL-12445) cells, as shown in<br />

Figure 1.<br />

The feeding of both runs was controlled only through the permittivity<br />

signal in order to maintain the asparagine concentration at a certain<br />

level. Asparagine was chosen due to its central role in cell metabolism<br />

and to the fact that it is usually a limiting substrate in CHO DP-12<br />

cultures. These proof-of-concept runs demonstrated that permittivitybased<br />

automated feeding can be a valuable tool for the optimization of<br />

fed-batch process parameters, such as feeding start, flow rate and<br />

composition.<br />

Conclusions: For suspension cultures with single cells and high viability a<br />

linear correlation (R 2 ≥ 0.98) of the permittivity signal with the viable cell<br />

density measured offline was obtained, at least for the exponential<br />

growth phase. Based on this correlation, a closed-loop controlled cell<br />

bleed was implemented in a perfusion process in which the permittivity


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P30) Totalviablecellcount,viablecelldensityandasparagineconcentration of the two proof-of-concept CHO DP-12 fed-batch<br />

processes (initial working volume: 1 L; 37°C; 40% DO; pH 7.1).<br />

signal was used to keep the viable cell density at a constant level of 10 7<br />

viable cells/mL. Furthermore, a linear correlation with the i-Biomass 465<br />

signal was observed for several substrates independent of the correlation<br />

between viable cell density and permittivity. Based on these results, a<br />

closed-loop controlled feeding was successfully established resulting in a<br />

fully automated fed-batch process.<br />

Acknowledgements: We would like to thank FOGALE nanotech for<br />

providing the i-Biomass 465 system and TeutoCell AG for supplying the<br />

cell culture media and feed solutions.<br />

References<br />

1. Boulton CA, Maryan PS, Loveridge D: The application of a novel biomass<br />

sensor to the control of yeast pitching rate. Proc 22nd Eur Brew Conv<br />

Zurich European Brewing Convention Oxford: Oxford University Press 1989,<br />

653-661.<br />

2. Noll T, Biselli M: Dielectric spectroscopy in the cultivation of suspended<br />

and immobilized hybridoma cells. J Biotechnol 1998, 63:187-198.<br />

3. Ansorge S, Esteban G, Schmid G: Multifrequency permittivity<br />

measurements enable on-line monitoring of changes in intracellular<br />

conductivity due to nutrient limitations during batch cultivations of<br />

CHO cells. Biotechnol Prog 2010, 26:272-283.<br />

P31<br />

CHO cell lines generated by PiggyBac transposition<br />

Mattia Matasci 1 , Virginie Bachmann 1 , Lucia Baldi 1 , David L Hacker 1 ,<br />

Maria De Jesus 2 , Florian M Wurm 1,2*<br />

1<br />

Laboratory of Cellular Biotechnology, Faculty of Life Sciences, Ecole<br />

Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland;<br />

2<br />

ExcellGene SA, CH-1870 Monthey, Switzerland<br />

E-mail: florian.wurm@epfl.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P31<br />

A major bottleneck in the manufacture of recombinant therapeutic proteins<br />

is the time and effort needed for the generation of stable, high-producing<br />

mammalian cell lines. Conventional gene transfer methods for stable cell<br />

line generation rely on random transgene integration, resulting in<br />

unpredictable and highly variable levels of expression of the transgene in<br />

individual clones [1,2]. As a consequence, a large number of stably<br />

transfected cells must be analyzed to recover a few high-producing clones.<br />

Recently, we described the use of a PiggyBac (PB) transposon for the<br />

generation of high-producing mammalian cell lines [3]. The PB dual vector<br />

system consists of 1) a donor vector carrying an artificial transposon with a<br />

mammalian expression cassette for the recombinant transgene and<br />

puromycin selection marker and 2) a helper vector driving transient<br />

expression of the PB transposase (PBase). PB transposition mediates stable<br />

transgene integration via a “cut and paste” mechanism in which the PBase<br />

excises the artificial transposon sequence from the plasmid and catalyzes its<br />

Page 52 of 181<br />

insertion into the host cell genome. A main advantages of the PB system<br />

over conventional passive integration are an improved efficiency of<br />

transgene integration resulting in a more integration events and more<br />

stable clones. Furthermore, PB favors transgene integration into actively<br />

transcribed regions of the host genome [4]. The PB transposon has a high<br />

cargo capacity of up to 14 Kb and transposition results in the stable<br />

genomic integration of well-defined sequences, thus reducing the<br />

probability of integration of truncated, non-functional transgenes [5]. Finally,<br />

recent reports have demonstrated the feasibility of using the PB system to<br />

obtain persistent expression of multiple genes carried either on a single or<br />

on distinct donor vectors [6].<br />

We initially determined the efficiency of the PB system in the generation of<br />

stable lines using suspension adapted mammalian cells. CHO and HEK 293<br />

cells were co-transfected with an eGFP-bearing donor plasmid along with<br />

the PB helper plasmid. The cells were then grown in the absence of<br />

puromycin, and the percentage of GFP-expressing cells in each culture was<br />

determined by flow cytometry on a daily basis. By 21 days post-transfection,<br />

the remaining GFP-positive cells were assumed to be recombinant.<br />

Compared to conventional transfection of plasmid DNA, PB transposition<br />

resulted in an improvement in the efficiency of stable cell line generation up<br />

to 20-fold for both CHO and HEK 293 cells (Figure 1A).<br />

To further evaluate the PB system, CHO cells expressing a tumour necrosis<br />

factor receptor:Fc fusion protein (TNFR:Fc) were generated either by PBtransposition<br />

or by conventional transfection. Clonal cell lines were<br />

recovered following selection in 50 or 10 µg/mL puromycin for two weeks.<br />

Recovered lines were grown in suspension culture for 7 days in 24-well<br />

plates after which the medium was analyzed by ELISA to determine TNFR:Fc<br />

productivity. Transposition increased the frequency of high-producing<br />

clones in the transfected population (Figure 1B). To further characterized for<br />

the level and stability of transgene expression the original cell pools<br />

generated by PB transposition or conventional transfection, as well as the<br />

top 4 producers from each transfection were cultivated in the absence of<br />

selection in serum-free suspension culture, over a period of 16 or 14 weeks,<br />

respectively. When compared to clones and cell pools generated by<br />

conventional transfection, PB-derived cell lines and cell pools produced up<br />

to 4-fold more recombinant protein and had greater transgene expression<br />

stability (Figure 1C)<br />

In conclusion our results demonstrate that stable cell lines derived by PB<br />

transposition are efficiently generated and are more productive than cell<br />

lines generated by conventional transfection methods. Therefore, the PB<br />

system represents a valuable and practical alternative to standard<br />

plasmid transfection to efficiently generate cell clones with stable and<br />

enhanced transgene expression.<br />

Acknowledgements: This work was supported by the Ecole Polytechnique<br />

Fédérale de Lausanne and the CTI Innovation Promotion Agency of the<br />

Swiss Federal Department of Economic Affairs (n. 10203.1PFLS-LS) under a<br />

collaboration with ExcellGene SA (Switzerland).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P31) A) Enhanced stable integration efficiency in CHO and HEK 293 cells by PB transposition. B) Productivity analysis of CHO clonal cell<br />

lines sorted from cell pools generated by PB transposition (PB) or conventional transfection (TX). C) Analysis of the stability of TNFR:Fc expression over<br />

time and transgene copy number, for cell pools [PB(50), PB(10), TX(50), and TX(10)] and clonal lines generated by PB transposition (PB50 and PB10) or<br />

conventional transfection (TX50 and TX10).<br />

References<br />

1. Matasci M, et al: Recombinant therapeutic protein production in<br />

cultivated mammalian cells: current status and future prospects. Drug<br />

Discovery Today: Technologies 2009, 5(2-3):e37-e42.<br />

2. Wurm FM: Production of recombinant protein therapeutics in cultivated<br />

mammalian cells. Nat Biotechnol 2004, 22(11):1393-8.<br />

3. Matasci M, Baldi L, Hacker DL, Wurm FM: The PiggyBac transposon<br />

enhances the frequency of CHO stable cell line generation and yields<br />

recombinant lines with superior productivity and stability. Biotechnol<br />

Bioeng 2011, doi: 10.1002/bit.23167.<br />

4. Galvan DL, et al: Genome-wide mapping of PiggyBac transposon<br />

integrations in primary human T cells. J Immunother 2009, 32(8):837-44.<br />

5. Ding S, et al: Efficient transposition of the piggyBac (PB) transposon in<br />

mammalian cells and mice. Cell 2005, 122(3):473-83.<br />

6. Kahlig KM, et al: Multiplexed transposon-mediated stable gene transfer in<br />

human cells. Proc Natl Acad Sci U S A 2010, 107(4):1343-8.<br />

P32<br />

Transposon mediated co-integration and co-expression of transgenes in<br />

CHO-DG44 cells<br />

Sowmya Balasubramanian, Mattia Matasci, Lucia Baldi, David L Hacker,<br />

Florian M Wurm *<br />

Laboratory for Cellular Biotechnology (LBTC), Faculty of Life Sciences, École<br />

Polytechnique Fédérale de Lausanne CH-1015 Lausanne, Switzerland<br />

E-mail: florian.wurm@epfl.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P32<br />

Background: Transposon systems mediate stable integration of exogenous<br />

DNA elements into a host cell genome, and have been successfully used in<br />

mammalian cells for the generation of stable cell lines. The piggyBac (PB)<br />

Page 53 of 181<br />

transposon system has been shown to have several advantages over the<br />

other transposon system available [1-3]. It has also been shown to generate<br />

stable cell lines at significantly higher frequency than the conventional<br />

transfections [3]. Here, we investigated the efficiency of the piggyBac (PB)<br />

transposon to facilitate the co-expression of multiple artificial transposons,<br />

each bearing a single transgene and the puromycin resistance gene for<br />

selection. Green fluorescent protein (eGFP), red fluorescent protein (mKate),<br />

and a human IgG1 antibody were used as model proteins [4]. The effect of<br />

the stringency of selection on pool productivity was determined with<br />

increasing concentrations of puromycin. The duration of selection necessary<br />

for the generation of recombinant cell pools was also tested by selecting for<br />

a period of either 5 or 10 days.<br />

Materials and methods:<br />

CHO-DG44 transfection: Cells were transfected using linear 25 kDa<br />

polyethylenimine (PEI) (Polysciences, Eppenheim, Germany). All the<br />

transfections are done in a final volume of 10 mL. Transfected cultures<br />

were incubated at 37°C in 5% CO 2 and 85% humidity with agitation at<br />

180 rpm. The ratio of plasmid coding for Gene of Interest (GOI) to the<br />

plasmid coding for the transposase was kept constant at 9:1.<br />

Generation of pools and clones: For the generation of stable pools, two<br />

days post transfection the cells were seeded at a density of 5 x 10 5 cells/mL<br />

in ProCHO5 and puromycin. The cells were placed under selection pressure<br />

for 5 or 10 days. In case of a 10 day selection period, the puromycin<br />

concentration in the cultures was replenished on day 7 post transfection by<br />

seeding at a density of 5 x10 5 cells/mL in fresh ProCHO5 with puromycin.<br />

For productivity analysis the cells were seeded at a density of 3 x10 5 cells/<br />

mL and analyzed at day four.<br />

Stable clones were generated by limiting dilution of the pools. The<br />

productivity of the clones was analyzed after five days of culture.<br />

Analyses: A Guava EasyCyte microcapillary flow cytometer (Millipore) with<br />

excitation and emission wavelengths of 488 and 532 nm, respectively, was


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P32) Comparison of the A) %GFP positive cells and B) IgG1 productivity between pools generated using standard transfection (left)<br />

and PB transposition (right).<br />

used to measure EGFP-specific fluorescence. The IgG concentration in the<br />

culture medium was determined by sandwich ELISA as previously<br />

described [4].<br />

Results:<br />

PB Transposition significantly enhances co-expression of multiple<br />

genes from stable pools compared to standard transfection: Cells were<br />

co-transfected with three artificial transposons namely, eGFP and the heavy<br />

and light chains of a human IgG1 antibody along with the plasmid coding<br />

for the transposase. Cell populations recovered after selection showed<br />

30 % of GFP positive cells for standard transfections, whereas for pools<br />

generated by PB transposition, the percentage of cells producing GFP was<br />

up to 80%, which represent a 2.5 fold improvement (Fig. 1A). IgG1<br />

productivity up to 120 mg/L was achieved using transposition, whereas in<br />

standard transfections the highest titers obtained were only 5 mg/L,<br />

corresponding to a 24 fold improvement (Fig. 1B).<br />

No significant differences were observed in the percentage of GFP positive<br />

cells and IgG productivity in cell pools generated by transposition with 5 or<br />

10 days of selection. However, increased IgG productivity and % GFPpositive<br />

cells were observed with 10 days of selection in case of standard<br />

transfection (data not shown). This shows that a longer duration of selection<br />

pressure is necessary to generate pools by standard transfections compared<br />

to transposition. Furthermore, increased antibody productivity was observed<br />

with increasing puromycin concentration (Fig. 1).<br />

PB transposition strongly improved clonal productivity: Four different<br />

plasmids coding for (1) EGFP, (2) mKATE, (3) the light and (4) heavy chain<br />

genes of a human IgG1 antibody. Clones for both the standard and the<br />

transposition-based transfections were generated by limiting dilution of the<br />

cell pools after selection for 10 days. The IgG productivities of 65 clones of<br />

each standard transfection and transposition were analyzed. About 96% of<br />

the clones generated by the standard transfection were found to be lowproducing.<br />

However, more than 40% of clones generated by transposition<br />

produced more than 20 mg/L and about 12% of the clones were highproducers<br />

(Table 1).<br />

Conclusions: Based on the above results we conclude that the piggyBac<br />

transposon system provides an efficient method for the co-integration of<br />

multiple genes. The use of the PB transposon system for the co-<br />

Table 1(abstract P32) Distribution of clones based on<br />

their IgG productivity<br />

Productivity Range Standard Transfection Transposition<br />

60 mg/L 0% 13%<br />

Page 54 of 181<br />

integration of multiple genes generates a higher frequency of highproducing<br />

clones than standard transfection. The GFP-expressing cell<br />

population was larger and the volumetric antibody productivity of the<br />

pools were higher with higher stringency of selection. A selection period<br />

with puromycin of only 5 days was sufficient for the generation of pools<br />

from transposon mediated transfection.<br />

Acknowledgments: This work has been supported in part by the CTI<br />

Innovation Promotion Agency of the Swiss Federal Department of Economic<br />

Affairs (n. 10203.1PFLS-LS), in collaboration with the company ExcellGene SA<br />

in Monthey, Switzerland.<br />

References<br />

1. Kahlig KM, Saridey AK, Kaja A, Daniels MA, George AL Jr, Wilson MH:<br />

Multiplexed transposon-mediated stable gene transfer in human cells.<br />

Proc Natl Acad of Sci USA 2010, 107(4):1343-8.<br />

2. Wilson MH, Coates CJ, George AL Jr: PiggyBac Transposon-mediated Gene<br />

Transfer in Human Cells. Molecular Therapy 2007, 15:139-145.<br />

3. Matasci M, Baldi L, Hacker DL, Wurm FM: The PiggyBac Transposon<br />

Enhances the Frequency of CHO Stable Cell Line Generation and Yields<br />

Recombinant Lines with Superior Productivity and Stability. Biotechnol<br />

Bioeng 2011, doi: 10.1002/bit.23167.<br />

4. Meissner P, Pick H, Kulangara A, Chatellard P, Friedrich K, Wurm FM:<br />

Transient gene expression: recombinant protein production with<br />

suspension-adapted HEK293-EBNA cells. Biotechnol Bioeng 2001,<br />

75:197-203.<br />

P33<br />

The use of filler DNA for improved transfection and reduced DNA<br />

needs in transient gene expression with CHO and HEK cells<br />

Divor Kiseljak, Yashas Rajendra, Sagar S Manoli, Lucia Baldi, David L Hacker,<br />

Florian M Wurm *<br />

Laboratory for Cellular Biotechnology, Faculty of Life Sciences, École<br />

Polytechnique Fédéral de Lausanne, CH-1015 Lausanne, Switzerland<br />

E-mail: florian.wurm@epfl.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P33<br />

Background: Transient gene expression (TGE) is a rapid method for the<br />

production of recombinant proteins. Protein productivity in TGE has<br />

improved significantly over the past decade, reaching 300 mg/L and 1 g/L in<br />

CHO DG44 (CHO) and HEK 293E (HEK) cells, respectively [1,2]. However, the<br />

amount of plasmid DNA needed for transfection remains relatively high,<br />

contributing significantly to the overall cost of the TGE process. In order to<br />

reduce the amount of plasmid DNA in TGE, we examined the possibility of<br />

partially replacing it with herring sperm DNA (non-coding “filler” DNA) in<br />

transfections of CHO and HEK cells.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Materials and methods:<br />

Transfections: Suspension-adapted CHO were centrifuged and<br />

resuspended in ProCHO5 (Lonza, Verviers, Belgium) at a density of 4 x<br />

10 6 cells/mL. Transfections of 5 mL were performed in TubeSpin® 50<br />

bioreactors (TPP, Trasadingen, Switzerland) using 0.625 µg DNA/1x10 6<br />

cells and 2.5 µg/1x10 6 cells of linear 25 kDa polye-thyleneimine (PEI;<br />

Polysciences, Eppenheim, Germany). pA3 carrying the genes for a human<br />

IgG light and heavy chains was used for transfections [1]. The transfected<br />

cultures were incubated at 31 °C in 5% CO2 and 85% humidity with<br />

agitation at 180 rpm. HEK cells were centrifuged and resuspended at a<br />

density of 20 x 10 6 cells/mL in RPMI 1640 medium (Lonza). To each<br />

culture, 1.0 µg DNA/1x10 6 cells and 3.0 µg PEI/1x10 6 cells were added. At<br />

3 h post-transfection, cells were diluted with Ex-Cell293 TM medium<br />

(Sigma) medium to a density of 1 x 10 6 cells/mL, and valproic acid was<br />

added to a final concentration of 3.75 mM. The transfected cultures were<br />

incubated at 37 °C as above.<br />

Analyses: The IgG concentration was determined by sandwich ELISA [3].<br />

To quantify plasmid DNA copy number, total cellular DNA was extracted<br />

using DNeasy Blood & Tissue Kit (Qiagen) according to the manufacturer’s<br />

protocol. To estimate the mRNA transgene levels, total RNA was extracted<br />

from cells using the GenElute mRNA kit (Sigma) according to the<br />

manufacturer’s protocol. DNA-free RNA was reverse transcribed using M-<br />

MLV reverse transcriptase (Sigma) and oligo dT as the primer. The RTqPCR<br />

was carried out in a LightCycler 480 Real-Time PCR System (Roche<br />

Applied Science, Basel, Switzerland) with the ABsolute QPCR SYBR Green<br />

ROX mix (Thermo Fisher Scientific) according to the manufacturer’s<br />

instructions.<br />

Results:<br />

Filler DNA allows considerable reduction in coding pDNA amounts:<br />

We tested the efficiency of herring sperm DNA as filler for TGE in CHO<br />

and HEK cells. We reduced the amount of pA3 to 17% or 33% of the<br />

optimum amount for each cell line and added filler DNA to 100%. The<br />

total amount of PEI was kept constant for all conditions. We observed<br />

that antibody titers increased when filler DNA was co-transfected with<br />

pA3 as compared to transfection with a reduced amount of pA3 alone<br />

(Fig. 1). These results showed that up to 83% of the coding pDNA could<br />

be replaced by filler DNA with only a minimal negative impact on yield.<br />

Filler DNA does not influence the delivery of coding pDNA: We<br />

investigated whether the use of filler DNA could improve pDNA delivery<br />

and/or intracellular pDNA stability by quantifying the plasmid copy<br />

number by qRT-PCR. The results showed that the plasmid copy number<br />

decreased proportionally with the amount of pA3 transfected in the<br />

presence or absence of filler DNA in both CHO and HEK cells (data not<br />

shown). Therefore, filler DNA did not influence the delivery of coding<br />

pDNA to transfected cells.<br />

Filler DNA enhances transgene mRNA levels and improves release<br />

of coding pDNA: We tested the hypothesis that the use of filler DNA<br />

could influence transcriptional competence of coding pDNA. By qPCR, we<br />

found that transgene mRNA levels were significantly higher in the<br />

presence of filler than in its absence (data not shown). This may explain<br />

the improved protein titers observed in the presence of filler DNA. We<br />

then hypothesized that filler DNA could influence the strength of PEI:DNA<br />

complexes and pDNA release from the complex upon delivery. With an in<br />

vitro dextran sulfate displacement assay we observed that with increasing<br />

Page 55 of 181<br />

PEI:DNA ratios, complex strength increased. However, when filler DNA<br />

was added to the complex, the release of pDNA from the complex was<br />

improved (data not shown).<br />

Conclusions: Our data show that in TGE the amount of the coding vector<br />

could be reduced considerably by replacement of a significant proportion<br />

of pDNA with filler DNA (herring sperm DNA) without a major negative<br />

impact on recombinant protein productivity. However, filler DNA did not<br />

influence the delivery or stability of pDNA. The addition of filler DNA to the<br />

DNA-PEI complex, however, relaxed the complex in vitro. Basedonthese<br />

results, we speculate that the presence of filler DNA results in a more<br />

efficient intracellular release of the pDNA from the DNA-PEI complex and<br />

thus to improved transgene transcription.<br />

Acknowledgments: This work has been supported in part by the CTI<br />

Innovation Promotion Agency of the Swiss Federal Department of<br />

Economic Affairs (n. 10563.1PFLS-LS) under a collaboration with<br />

ExcellGene SA (Monthey, Switzerland). TPP (Trasadingen, Switzerland) is<br />

acknowledged for providing TubeSpin® bioreactor 50 tubes.<br />

References<br />

1. Rajendra Y, Kiseljak D, Baldi L, Hacker D, Wurm FM: A simple high-yielding<br />

process for transient gene expression in CHO cells. J Biotechnol 2011,<br />

153:22-26.<br />

2. Backliwal G, Hildinger M, Chenuet S, Wulhfard S, De Jesus M, Wurm FM:<br />

Rational vector design and multi-pathway modulation of HEK 293E cells<br />

yield recombinant antibody titers exceeding 1 g/l by transient<br />

transfection under serum-free conditions. Nuc Acid Res 2008, 36(15).<br />

3. Meissner P, Pick H, Kulangara A, Chatellard P, Friedrich K, Wurm FM: Transient<br />

gene expression: recombinant protein production with suspensionadapted<br />

HEK293-EBNA cells. Biotechnol Bioeng 2001, 75:197-203.<br />

P34<br />

Rapid recombinant protein production from pools of transposongenerated<br />

CHO cells<br />

Mattia Matasci 1 , Virginie Bachmann 1 , Lucia Baldi 1 , David L Hacker 1 ,<br />

Maria De Jesus 2 , Florian M Wurm 1,2*<br />

1<br />

Laboratory of Cellular Biotechnology, Faculty of Life Sciences, Ecole<br />

Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland;<br />

2<br />

ExcellGene SA, CH-1870 Monthey, Switzerland<br />

E-mail: florian.wurm@epfl.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P34<br />

Transient gene expression (TGE) is the most commonly used technology for<br />

the rapid production of moderate quantities of recombinant proteins for<br />

preclinical studies or for analytical assay development [1,2]. Whereas TGE can<br />

provide milligram to gram amounts of recombinant proteins within a time<br />

period as short as few days, technical limitations and high costs of plasmid<br />

DNA hamper the application of this technology at larger scales. Here we<br />

describe an alternative method for the fast production of recombinant<br />

proteins from pools of mammalian cells stably transfected with the PiggyBac<br />

(PB) transposon. Expression from stably integrated transgene is highly<br />

dependent upon the chromatin structure surrounding the site of integration<br />

[3]. As a consequence stable cell populations generated by conventional<br />

transfection techniques generally show low productivity and reduced<br />

expression stability. Several studies showed that the PB transposase mediated<br />

Figure 1(abstract P33) Effect of filler DNA on transient IgG production in A) CHO and B) HEK cells. IgG titers were measured on day 7 post-transfection<br />

by ELISA. The DNA amounts are presented in μg/10 6 cells.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P34) Analysis of the stability or recombinant protein expression in 5 independent pools of CHO cells<br />

CHO-Pool Days post transfection % eGFP positive cells (*)<br />

TNFR:Fc productivity (mg/L) (°)<br />

30 92.8 ± 2.1 430 ± 46<br />

1 60 93.3 ± 0.2 455 ±10<br />

90 92.5 ± 1.2 473 ± 10<br />

30 90.0 ± 0.8 348 ± 36<br />

2 60 91.4 ± 0.5 370 ± 8<br />

90 94.0 ± 1.1 325 ± 35<br />

30 94.0 ± 1.5 432 ± 25<br />

3 60 94.2 ± 1.1 451 ± 28<br />

90 91.9 ± 1.8 494 ± 16<br />

30 92.0 ± 0.2 432 ± 30<br />

4 60 90.1 ± 2.0 407 ± 23<br />

90 92.3 ± 1.3 406 ± 28<br />

30 96.1 ± 1.5 388 ± 52<br />

5 60 92.4 ± 1.4 466 ± 19<br />

90 93.6 ± 0.2 372 ± 12<br />

(*) determined by GUAVA flow cytometry; (°) determined by ELISA.<br />

Page 56 of 181<br />

Figure 1(abstract P34) (A) Schematic representation of the protocol for the rapid production of recombinant proteins from pools of transposed cells. This<br />

protocol was successfully used to produce a recombinant monoclonal antibody (B) and two variants of TNFR:Fc (C and D). For each bioprocess shown, the<br />

percentage of viable cells (dotted lines) the viable cell density (dashed lines), and the recombinant protein titer (solid lines) were measured at the timesindicated.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

transgene insertion predominantly into actively transcribed regions of the<br />

mammalian genome [4,5], moreover we recently demon-strated that PB<br />

transposition generates high-producing CHO cell lines at a higher frequency<br />

than conventional plasmid transfection [6]. These results prompted us to<br />

develop a technology based on the use of stable pools generated by PB<br />

transposition for the rapid and scalable production of recombinant proteins.<br />

Initial experiments were aimed at determining the proportion of transgene<br />

expressing cells as well as the level and stability of recombinant protein<br />

production in PB transposed pools. First, we analyzed transgene expression in<br />

952 cell lines recovered from a pool of CHO cells generated by PBtransposition<br />

to express tumor necrosis factor receptor as an Fc fusion protein<br />

(TNFR:Fc). The cell lines were analyzed by ELISA for TNFR:Fc expression in 4day<br />

batch cultures. The clones showed levels of TNFR:Fc expression up to 360<br />

mg/L with an overall mean of 96 +/- 54 mg/L. PB-transposition resulted in a<br />

high percentage (more than 98%) of TNFR:Fc expressing clones (data not<br />

shown). Analyses on the stability of transgene expression over time were<br />

conducted using a bicistronic PB-donor plasmid allowing co-expression of<br />

TNFR:Fc and the enhanced green fluorescent protein (eGFP). Five<br />

independent pools of cells were generated by transposition and recovered<br />

after 10 days of selection in puromycin. The pools were cultivated for 3<br />

months in the absence of selection and eGFP expression was monitored<br />

periodically by flow cytometry. For each pool, the percentage of eGFPpositive<br />

cells remained stable over time (Table 1). The stability of transgene<br />

expression was further confirmed by TNFR:Fc productivity studies performed<br />

in 50-ml cultures at different time points post-transfection. At one month<br />

post-transfection, the five pools showed comparable growth and production<br />

characteristics, reaching TNFR:Fc titers in the range of 350-500 mg/L in 14-day<br />

batch cultures. Similar results were obtained when productivity was tested<br />

2 – 3 months post-transfection (Table 1).<br />

We developed a protocol for protein production from transposed cell pools<br />

at the 0.5-L scale (Figure 1A). Starting at 2 d post-transfection cells were<br />

subjected to 10 days of puromycin selection during which cells were<br />

expanded from TubeSpin® Bioreactor 50 tubes into orbitally shaken 250-mL<br />

cylindrical bottles. The batch bioprocess was finally started at 12 d posttransfection<br />

using orbitally shaken TubeSpin® Bioreactor 600 tubes. Using<br />

stable cell pools expressing either an IgG antibody (Fig. 1B) or two TNFR:Fc<br />

variants (Fig. 1C, D), we produced 500-750 mg of recombinant protein<br />

within a month after transfection.<br />

Our results demonstrated an improved level and stability of transgene<br />

expression in transposed pools, indicating usefulness of PB transposed<br />

cell pools as a valuable alternative to TGE for the rapid production of<br />

recombinant proteins.<br />

Acknowledgements: This work was supported by the Ecole Polytechnique<br />

Fédérale de Lausanne and the CTI Innovation Promotion Agency of the<br />

Swiss Federal Department of Economic Affairs (n. 10203.1PFLS-LS) under<br />

collaboration with ExcellGene SA (Switzerland).<br />

References<br />

1. Baldi L, Hacker DL, Adam M, Wurm FM: Recombinant protein production<br />

by large-scale transient gene expression in mammalian cells: state of<br />

the art and future perspectives. Biotechnol Lett 2007, 29(5):677-84.<br />

2. Hacker DL, De Jesus M, Wurm FM: 25 years of recombinant proteins from<br />

reactor-grown cells - where do we go from here? Biotechnol Adv 2009,<br />

27(6):1023-7.<br />

3. Kwaks TH, Otte AP: Employing epigenetics to augment the expression of<br />

therapeutic proteins in mammalian cells. Trends Biotechnol 2006, 24(3):137-42.<br />

4. Ding S, et al: Efficient transposition of the piggyBac (PB) transposon in<br />

mammalian cells and mice. Cell 2005, 122(3):473-83.<br />

5. Galvan DL, et al: Genome-wide mapping of PiggyBac transposon<br />

integrations in primary human T cells. J Immunother 2009, 32(8):837-44.<br />

6. Matasci M, Baldi L, Hacker DL, Wurm FM: The PiggyBac transposon<br />

enhances the frequency of CHO stable cell line generation and yields<br />

recombinant lines with superior productivity and stability. Biotechnol<br />

Bioeng 2011, doi: 10.1002/bit.23167.<br />

P35<br />

Influence of glutamine on transient and stable recombinant protein<br />

production in CHO and HEK-293 cells<br />

Yashas Rajendra, Divor Kiseljak, Lucia Baldi, David L Hacker, Florian M Wurm *<br />

Laboratory for Cellular Biotechnology (LBTC), École Polytechnique Fédéral de<br />

Lausanne (EPFL), CH-1015 Lausanne, Switzerland<br />

E-mail: florian.wurm@epfl.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P35<br />

Page 57 of 181<br />

Background: Glutamine is an essential component in culture media for<br />

most of the mammalian cell lines. It is often used as an alternative source of<br />

energy by cells, along with glucose. Glutamine metabolism induces<br />

ammonia accumulation in cell culture. Elevated ammonia concentration<br />

above 2 mM has been shown to have negative impact on both cell growth<br />

and recombinant protein productivity [1-4]. In this study we investigated the<br />

effects of decreased glutamine concentration in the medium for CHO-DG44<br />

and HEK-293E cells during transient gene expression (TGE). The rationale<br />

was to reduce ammonia accumulation in the culture, and consequently,<br />

improve cell viability and recombinant protein productivity.<br />

Materials and methods:<br />

CHO-DG44 transfection: The cells were centrifuged and resuspended in<br />

ProCHO5 medium (Lonza, Verviers, Belgium) at a density of 5x10 6 cells/mL<br />

in orbitally shaken 250 ml glass bottles. Each transfection was performed<br />

in 100 mL of culture using 0.6 µg of plasmid DNA and 3.0 µg of linear<br />

25 kDa polyethyleneimine (PEI, Polysciences, Eppelheim, Germany; pH 7)<br />

per million cells. The transfected cultures were incubated at 31 °C in 5%<br />

CO2 and 85% humidity with agitation at 120 rpm.<br />

HEK-293E transfection: The cells were centrifuged and resuspended at<br />

density of 20x10 6 cells/mL in RPMI 1640 medium. Each transfection was<br />

performed in 100 mL of culture using 1.5 µg of plasmid DNA and 3.0 µg of<br />

linear 25 kDa PEI per 10 6 cells. Three hours post-transfection, cells were<br />

diluted with Ex-Cell293 medium (Sigma, Saint-Louis, USA) to a density of<br />

2x10 6 cells/mL, and valproic acid was added to a final concentration of<br />

3.75 mM. The transfected cultures were incubated at 37 °C in 5% CO 2 and<br />

85% humidity with agitation at 120 rpm.<br />

Stable Clone and Pool: A cell line and cell pool expressing anti-Rhesus<br />

IgG was kindly provided by Tatiana Benavides from our lab. Cell line was<br />

established by transfection of plasmid containing both light and heavy<br />

chain into CHO-DG44 cells, followed by flow cytometer sorting and<br />

limiting dilution. Cell pool was established by transfection of plasmid<br />

containing both light and heavy chain into CHO-DG44 cells, followed by<br />

selection under puromycin for two weeks.<br />

Metabolic analytes and IgG levels: The levels of glucose, glutamine,<br />

ammonium, and lactate were determined with a BioProfile 200 Bioanalyzer<br />

(Nova Biomedical Corp., Waltham, MA). The IgG concentration in the<br />

culture medium was determined by sandwich ELISA as previously<br />

described [5].<br />

Results:<br />

Low glutamine concentration improved transient IgG production:<br />

Different concentrations of free glutamine in medium, ranging from 0 mM<br />

to 6 mM were tested. Both cell lines showed improved production of IgG<br />

with a reduced glutamine concentration (Fig. 1 panel A). The optimal<br />

concentration of glutamine in terms of IgG production was 2 mM for CHO-<br />

DG44 cells and 0 mM for HEK-293E cells. We observed a 50% improvement<br />

in IgG production for both CHO-DG44 and HEK293 cells. We did not<br />

observe a significant difference on cell density or viability between the<br />

tested concentrations of glutamine for both CHO-DG44 and HEK293 cells<br />

during the entire course of the culture (data not shown).<br />

Higher initial concentration of glutamine resulted in higher concentration<br />

of ammonia, up to 5 mM for the highest glutamine concentration tested<br />

(Fig 1 panel B).<br />

Lactate accumulation in both CHO-DG44 and HEK-293E cells was observed<br />

during the initial phase of the cultures, while the cells were actively<br />

growing. In CHO-DG44 cells, consumption of lactate started immediately<br />

after its accumulation. In HEK-293E cells, lactate levels remained either at a<br />

constant level or, in the absence of glutamine, continued to increase over<br />

the entire culture (data not shown). Glucose and glutamine consumption<br />

rate remained unaffected under all the conditions tested.<br />

Lower initial glutamine concentration improved stable IgG production<br />

in growth-arrested stable CHO-DG44 cells and pools: A stable cell clone<br />

and cell pool of CHO-DG44 cells expressing IgG were cultivated in the<br />

presence of different glutamine concentrations under mild hypothermia<br />

conditions (31 °C). Both the clone and the cell pool showed improved<br />

production of IgG with reduction in glutamine concentration. The IgG titers<br />

were approximately 80% higher for the clone and 60% higher for the pool<br />

at 0 mM glutamine, compared to titers obtained with 6 mM glutamine<br />

(data not shown).<br />

Conclusions: The effects of different concentrations of glutamine on IgG<br />

production in growth arrested cells were investigated. Recently published<br />

results show that CHO-DG44 cells are arrested in G1 phase of the cell cycle<br />

under mild hypothermia at 31 °C [6]. For HEK-293E cells growth arrest was<br />

induced with VPA [7]. We conclude that a lower glutamine concentration


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P35) Effect of glutamine concentration on relative A) IgG production and B) Ammonia accumulation in CHO-DG44 cells and HEK-293E<br />

cells under transient transfection conditions on day 7.<br />

results in improved transient antibody titers in CHO-DG44 and HEK-293E<br />

cells mainly due to lower accumulation of ammonia in the culture, which<br />

has previously been shown to have a negative impact on cellular<br />

productivity [1-4]. Glutamine reduction also had a positive impact on<br />

recombinant IgG production in a stable clone and a pool of recombinant<br />

CHO-DG44 cells at 31°C. These data suggest that this strategy may be<br />

successful in both transient and stable gene expression processes under<br />

conditions of growth arrest.<br />

Acknowledgments: We thank Tatiana Benavides and Mattia Matasci for<br />

providing the CHO clone and pool. Part of this work was supported by the<br />

Swiss Innovation Promotion Agency KTI/CTI of the Swiss Federal Department<br />

of Economic Affairs (n. 10203.1PFLS-LS) under a collaboration with<br />

ExcellGene SA (Switzerland).<br />

References<br />

1. Canning WM, Fields BN: Ammonium chloride prevents lytic growth of<br />

reovirus and helps to establish persistent infection in mouse L cells.<br />

Science 1983, 219:987-988.<br />

2. Hansen HA, Emborg C: Influence of ammonium on growth, metabolism,<br />

and productivity of a continuous suspension Chinese hamster ovary cell<br />

culture. Biotechnol Prog 1994, 10:121-124.<br />

3. Ito M, McLimans WF: Ammonia inhibition of interferon synthesis. Cell Biol<br />

Int Rep 1981, 5:661-666.<br />

4. Reuveny S, Velez D, Macmillan JD, Miller L: Factors affecting cell growth<br />

and monoclonal antibody production in stirred reactors. J Immunol<br />

Methods 1986, 86:53-59.<br />

5. Meissner P, Pick H, Kulangara A, Chatellard P, Friedrich K, Wurm FM:<br />

Transient gene expression: recombinant protein production with<br />

suspension-adapted HEK293-EBNA cells. Biotechnol Bioeng 2001,<br />

75:197-203.<br />

6. Wulhfard S, Tissot S, Bouchet S, Cevey J, De Jesus M, Hacker DL, Wurm FM:<br />

Mild hypothermia improves transient gene expression yields several fold<br />

in Chinese hamster ovary cells. Biotechnol Prog 2008, 24:458-465.<br />

7. Greenblatt DY, Vaccaro AM, Jaskula-Sztul R, Ning L, Haymart M,<br />

Kunnimalaiyaan M, Chen H: Valproic acid activates notch-1 signaling and<br />

regulates the neuroendocrine phenotype in carcinoid cancer cells.<br />

Oncologist 2007, 12:942-951.<br />

P36<br />

kLa as a predictor for probe-independent mammalian cell bioprocesses<br />

in orbitally shaken bioreactors<br />

Stéphanie Tissot, Dominique T Monteil, Lucia Baldi, David L Hacker,<br />

Florian M Wurm *<br />

Laboratory of Cellular Biotechnology, Faculty of Life Sciences, Ecole<br />

Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland<br />

E-mail: florian.wurm@epfl.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P36<br />

Page 58 of 181<br />

Background: Orbitally shaken flasks are commonly used at an early stage<br />

of bioprocess development with mammalian cells. In contrast to large-scale<br />

stirred-tank bioreactors, shaken flasks are usually operated in probeindependent<br />

bioprocesses, i.e. without strictly controlling the pH or<br />

dissolved oxygen concentration (DO). As a consequence, gas transfer issues<br />

are thought to limit the effectiveness of orbitally shaken flasks and<br />

bioreactors (OSRs). To define optimal operating conditions for probeindependent<br />

bioprocesses in OSRs, we tested the effects of the mass<br />

transfer coefficient of oxygen (k La) on mammalian cell growth, recombinant<br />

protein production, and environmental conditions of the culture (pH, DO).<br />

Materials and methods: The k La was measured by the dynamic method<br />

describedin[1]usingnon-invasiveO 2 sensors (PreSens, Regensburg,<br />

Germany). A recombinant CHO DG44-derived cell line expressing a human<br />

IgG monoclonal antibody (CHO-IgG) [3] was cultivated in suspension as<br />

described [4]. To investigate the effects of the k La on cell growth, CHO-IgG<br />

cells were into 1-L cylindrical bottles with working volumes from 200 to 600<br />

mL. The bottles were equipped with vented caps and orbitally shaken at<br />

110 rpm in an incubator at 37°C with 5% CO 2. To test the k La as a scale-up<br />

factor, CHO-IgG cells were inoculated at 0.3 million cells/mL in a 200-L OSR<br />

(Kühner AG, Birsfelden, Switzerland) with a working volume of 100 L and<br />

agitated at 57 rpm. Air containing 5% CO 2 was flushed into the OSR at 1 L<br />

min -1 . After overnight cultivation, samples were withdrawn from the 100-L<br />

culture and used to inoculate satellite cultures in 1- and 5-L bottles with<br />

vented caps. The volume of the cultures in bottles was adjusted to obtain<br />

the same k La as the one in the 200-L bioreactor (7 h −1 ), and the bottles were<br />

agitated at 110 rpm.<br />

Results: In a 1-L OSR the k La decreased from 11 to 3 h-1 as the working<br />

volume increased from 200 to 600 mL (Fig. 1a). As the working volume of<br />

the cultures increased in the 1-L OSR, the DO decreased (Fig. 1b). In all the<br />

cultures, the pH decreased with time of cultivation (Fig. 1c) At working<br />

volumes greater than 400 mL (k La


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

CHO-IgG cells. Cell cultivation in a 200-L OSR without pH or DO controllers<br />

resulted in similar cell densities, recombinant protein titers and pH values<br />

as in 1- and 5-L OSRs when the three types of OSRs were operated at the<br />

same k La. These results suggest that large-scale bioprocesses can be<br />

operated without pH or DO controllers as long as a sufficient kLa is<br />

maintained through appropriate cultivation conditions (e.g. working<br />

volume, agitation rate, geometry of the vessel).<br />

Acknowledgments: We thank Dr. Mattia Matasci for providing the CHO-<br />

IgG cell line. We gratefully acknowledge: Kühner AG, and Sartorius-Stedim<br />

Biotech, for the considerable support of equipment and material. This work<br />

has been supported by the CTI Innovation Promotion Agency of the Swiss<br />

Federal Department of Economic Affairs and by the Swiss National Science<br />

Foundation (SNSF).<br />

References<br />

1. Gupta A, Rao G: A study of oxygen transfer in shake flasks using a noninvasive<br />

oxygen sensor. Biotechnol Bioeng 2003, 84:351-358.<br />

2. Oberbek A, Matasci M, Hacker DL, Wurm FM: Generation of stable, highproducing<br />

CHO cell lines by lentiviral vector-mediated gene transfer in<br />

serum-free suspension culture. Biotechnol Bioeng 2011, 108:600-610.<br />

3. Matasci M, Baldi L, Hacker DL, Wurm FM: The PiggyBac transposon<br />

enhances the frequency of CHO stable cell line generation and yields<br />

Page 59 of 181<br />

Figure 1(abstract P36) Effects of the k La on CHO-IgG cell cultures. The k La was measured in 1-L OSR with working volumes from 200 to 600 mL (a). The<br />

CHO-IgG cells were cultivated in 1-L OSR in 200 (⃝), 300 (⃞), 400 (Δ), 500 (●) and 600 mL (■). The DO (b), pH (c) and viable cell density (d) were<br />

measured at the times indicated. The shaking diameter was 5 cm.<br />

recombinant lines with superior productivity and stability. Biotechnol<br />

Bioeng 2011, DOI: 10.1002/bit.23167.<br />

4. Muller N, Girard P, Hacker DL, Jordan M, Wurm FM: Orbital shaker<br />

technology for the cultivation of mammalian cells in suspension.<br />

Biotechnol Bioeng 2005, 89:400-406.<br />

P37<br />

Transient transfection of insect Sf-9 cells in TubeSpin® bioreactor<br />

50 tubes<br />

Xiao Shen 1† , Patrik O Michel 1† , Qiuling Xie 1,2 , David L Hacker 1 ,<br />

Florian M Wurm 1*<br />

1<br />

Laboratory of Cellular Biotechnology, Faculty of Life Sciences, Ecole<br />

Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland;<br />

2<br />

Institute of Bioengineering, Jinan University, Guangzhou 510632,<br />

Guangdong, P. R. China<br />

E-mail: florian.wurm@epfl.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P37<br />

Background: Sf-9 cells, derived from Spodoptera frugiperda, arewidely<br />

used for recombinant protein production using the baculovirus expression


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

vector system (BEVS). However, this results in a productive viral infection<br />

and cell lysis. Therefore, a non-lytic, plasmid-based expression system for<br />

suspension Sf-9 cells would be a valuable alternative to the BEVS for rapid,<br />

scalable, and high-yielding recombinant protein production and for the<br />

generation of stable Sf-9 cell lines [4]. In this work, we present a simple,<br />

efficient and cost-effective plasmid-based method for transient expression<br />

of recombinant proteins in Sf-9 cells cultivated in serum-free suspension<br />

mode in a high-throughput culture system, TubeSpin® bioreactor 50 tubes<br />

(TubeSpins).<br />

Materials and methods: Sf-9 cells were maintained in suspension in<br />

TubeSpins (TPP, Trasadingen, Switzerland) at 28 °C [5]. The human tumor<br />

necrosis factor receptor-Fc fusion protein (TNFR-Fc) gene was cloned into<br />

pIEx10 (Novagen, Merck, Darmstadt, Germany) to generate pIEx-TNFR-Fc.<br />

The cells in exponential growth phase were inoculated in fresh Sf900 II<br />

medium (Invitrogen, Carlsbad, CA) one day prior to transfection. The next<br />

day, the cells were transfected with 1.5 µg pTNFR-Fc and 2.25 µg linear<br />

25 kDa polyethylenimine (PEI, Polysciences, Warrington, PA) per 10 6 cells.<br />

The DNA and PEI were first mixed in de-ionized water at room temperature<br />

for 10 min prior to addition to the culture. At the time of transfection the<br />

cell density was 20 x 10 6 cells per mL. Cells were subsequently diluted to 4 x<br />

10 6 cells per mL with fresh Sf900 II media to allow for growth. The culture<br />

was maintained at 28 °C in an incubator shaker for 7 d [5]. The TNFR-Fc<br />

concentration in the medium was determined by ELISA as described [6].<br />

Results: Sf-9 cells were transfected in TubeSpins with pIEx-TNFR-Fc. By 7 d<br />

post-transfection, the TNFR-Fc concentration reached 42 mg/L (Figure 1).<br />

In a separate transfection with a plasmid expression the enhanced green<br />

fluorescent protein (GFP) gene, 58% of the cells were GFP-positive at 5 d<br />

post-transfection (data not shown).<br />

Page 60 of 181<br />

Conclusions: This study validates the use of PEI for transient expression in<br />

suspension Sf-9 cell cultures. This system was used to produce both<br />

intracellular (GFP) and secreted (TNFR-Fc) proteins. The results show that<br />

PEI-mediated transient transfection is a fast and efficient alternative to BEVS<br />

for high-yielding protein expression in Sf-9 cells.<br />

Acknowledgements: This work has been supported by the CTI Innovation<br />

Promotion Agency of the Swiss Federal Department of Economic Affairs, by<br />

grants from the Guangdong Provincial Department of Science and<br />

Technology (2008B030301349), the MOE of China (211 Grant), and the<br />

Academy of Finland (decision no. 135820). TPP (Trasadingen, Switzerland) is<br />

acknowledged for providing TubeSpin® bioreactor 50 tubes.<br />

References<br />

1. De Jesus M, Wurm FM: Manufacturing recombinant proteins in kg-ton<br />

quantities using animal cells in bioreactors. Eur J Pharm Biopharm 2011,<br />

78:184-188.<br />

2. Harrison RL, Jarvis DL: Transforming lepidopteran insect cells for continuous<br />

recombinant protein expression. Methods Mol Biol 2007, 388:299-316.<br />

3. Jarvis DL: Baculovirus-insect cell expression systems. Methods Enzymol<br />

2009, 463:191-222.<br />

4. Li E, Brown SL, Dolman CS, Brown GB, Nemerow GR: Production of<br />

functional antibodies generated in a nonlytic insect cell expression<br />

system. Prot Expr Purif 2001, 21:121-128.<br />

5. Xie Q, Michel PO, Baldi L, Hacker DL, Zhang X, Wurm FM: TubeSpin<br />

bioreactor 50 for the high-density cultivation of Sf-9 insect cells in<br />

suspension. Biotechnol Lett 2011, 33:897-902.<br />

6. Oberbek A, Matasci M, Hacker DL, Wurm FM: Generation of stable, highproducing<br />

CHO cell lines by lentiviral vector-mediated gene transfer in<br />

serum-free suspension culture. Biotechnol Bioeng 2011, 108:600-610.<br />

Figure 1(abstract P37) Sf-9 cells were transfected with pTNFR-Fc (■) plasmid DNA and PEI . The TNFR-Fc concentration in the medium was determined<br />

by ELISA at the times indicated.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

P38<br />

Transient gene expression with CHO cells in conditioned medium:<br />

a study using TubeSpin® bioreactors<br />

João Pereira, Yashas Rajendra, Lucia Baldi, David L Hacker, Florian M Wurm *<br />

Laboratory of Cellular Biotechnology, Faculty of Life Sciences, Ecole<br />

Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland<br />

E-mail: florian.wurm@epfl.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P38<br />

Background: Transient gene expression (TGE) allows rapid protein<br />

production in mammalian cells and has become an important tool in the<br />

Page 61 of 181<br />

pharmaceutical product development pipeline [1]. Polyethylenimine (PEI)mediated,<br />

high-density transfection allowed to express recombinant<br />

proteins at yields exceeding 1 g/L in only a few weeks [2]. Although highly<br />

efficient protocols are available, volumetric scale-up of TGE is still a<br />

challenge. A major issue is the need to perform the transfection in fresh<br />

medium rather than in conditioned (spent) medium. This implies a<br />

medium exchange step just before transfection. In CHO-DG44 [3] cells we<br />

observed up to a 100-fold decrease in volumetric protein production if<br />

transfections were performed in conditioned medium, compared to fresh<br />

medium. The reasons for such a negative effect of conditioned medium on<br />

transfectability and/or protein production expression are not yet known.<br />

To study this problem we transfected CHO cells at small-scale in TubeSpin®<br />

Figure 1(abstract P38) Using the TubeSpin® bioreactors, 38 commercially available cell culture media were studied for their impact in cell growth and in<br />

transient protein production in a 7 day batch process at the 5 mL scale. Error < 5 % for growth rate. (n=2).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

bioreactor 50 tubes using 41 different commercially available serum-free<br />

media formulations in combination with different transfection parameters<br />

and culture conditions. By comparing the transient production of a<br />

recombinant IgG antibody among the different media, we observed<br />

variation of up to 400-fold when transfecting in fresh media and up to<br />

20-fold when using conditioned medium. The optimization of the PEI:DNA<br />

ratio allowed a significant improvement in yields of transfection in<br />

conditioned medium.<br />

Methods: Suspension-adapted CHO-DG44 cells [3] were routinely<br />

cultivated in TubeSpin® bioreactor 50 tubes (TPP, Trasadingen,<br />

Switzerland) in ProCHO5 medium (Lonza, Vervier, Belgium) supplemented<br />

with 13.6 mg/L hypoxanthine, 3.9 mg/L thymidine, and 4 mM glutamine.<br />

The 38 media samples for transfection were provided by Excellgene SA.<br />

Conditioned medium is defined as a cell culture medium where cells<br />

have been growing for more than two days up to a density between 4-5<br />

million cells/mL. Cell growth was accessed with the Packed Cell Volume<br />

method. The dual expression vector pXLGCHO-A3, containing the cDNAs<br />

coding for human anti-Rhesus D IgG1 heavy and light chain cloned in<br />

separate expression cassettes in a head-to-head orientation, was kindly<br />

provided by Excellgene SA [4]. Transfections are performed at a cell<br />

density of 5.5 million cells/mL at a volume of 5 mL by the direct addition<br />

of 15 µg of pDNA and 76 µg of linear 25 kDa Polyethylenimine (PEI,<br />

Polysciences, Eppenheim, Germany).<br />

Results: Cells were inoculated in 38 different commercially available serum<br />

free media formulations tested, and cell growth was assessed daily by the<br />

packed cell volume method [5], over 3 days of culture. We identified 16<br />

media formulations that allowed for fast cellular growth (doubling time<br />

< 20h) up to the cell density of transfection (Figure 1). The same TGE<br />

protocol was applied in the 38 media formulations tested in a 7-day long<br />

batch process. Antibody productivity was analyzed by ELISA at day 7.<br />

Recombinant IgG production was observed only in 13 among the 38<br />

media formulations studied, and only in 5 media titers over 100 mg/L were<br />

achieved. Transfection in conditioned media yielded in the best cases only<br />

1/10 th of the production titers observed in fresh medium. Interestingly, the<br />

chemically defined media which sustained the fastest cellular growth (1-5,<br />

Figure 1) were not suitable for transient gene expression under the<br />

transfection conditions applied in this study.<br />

In a separate study, we tested different transfection conditions that could<br />

lead to an improvement of the process yields (data not shown). We<br />

observed that increasing the PEI and DNA concentrations could improve<br />

transient gene expression titers 3-fold when transfecting in conditioned<br />

medium.<br />

Conclusion: This work shows that the cell culture medium has a strong<br />

impact on the transient gene expression process. We have observed that<br />

even in media formulations that sustain a very good cell growth,<br />

polycation-mediated transfection is not very efficient. When transfecting in<br />

conditioned medium with a transfection procedure optimized for fresh<br />

medium, the transient gene expression process is not satisfactory in any of<br />

the media formulation tested. We were able to improve the transfection<br />

process by changing the PEI:DNA ratio (data not shown). In order to design<br />

a robust transfection protocol that would be effective in fresh medium as<br />

well as in conditioned medium, it is necessary to understand what factors<br />

affect the transfection negatively in the latter. This study suggests that a<br />

process without a medium exchange can be designed, facilitating easier<br />

scale-up of transient gene expression.<br />

Acknowledgments: This work was supported by the Ecole Polytechnique<br />

Fédérale de Lausanne and the CTI Innovation Promotion Agency of the<br />

Swiss Federal Department of Economic Affairs (n. 10563.1PFLS-LS) under<br />

a collaboration with ExcellGene SA (Monthey, Switzerland). TPP<br />

(Trasadingen, Switzerland) is acknowledged for providing TubeSpin®<br />

bioreactor 50 tubes.<br />

References<br />

1. Baldi L, Hacker DL, Adam M, Wurm FM: Recombinant protein<br />

production by large-scale transient gene expression in mammalian<br />

cells: state of the art and future perspectives. Biotechnol Lett 2007,<br />

29(5):677-84.<br />

2. Geisse S: Reflections on more than 10 years of TGE approaches. Protein<br />

Expr Purif 2009, 64(2):99-107.<br />

3. Urlaub G, Chasin LA: Isolation of Chinese hamster cell mutants deficient<br />

in dihydrofolate reductase activity. Proc Natl Acad Sci U S A 1980,<br />

77(7):4216-20.<br />

Page 62 of 181<br />

4. Miescher S, Zahn-Zabal M, De Jesus M, et al: CHO expression of a novel<br />

human recombinant IgG1 anti-RhD antibody isolated by phage display.<br />

Br J Haematol 2000, 111(1):157-66.<br />

5. Stettler M, Jaccard N, Hacker D, et al: New disposable tubes for rapid and<br />

precise biomass assessment for suspension cultures of mammalian cells.<br />

Biotechnol Bioeng 2006, 95(6):1228-33.<br />

P39<br />

Hydrodynamic stress in orbitally shaken bioreactors<br />

Stéphanie Tissot 1 , Martino Reclari 2 , Samuel Quinodoz 3 , Matthieu Dreyer 2 ,<br />

Dominique T Monteil 1 , Lucia Baldi 1 , David L Hacker 1 , Mohamed Farhat 2 ,<br />

Marco Discacciati 3 , Alfio Quarteroni 3 , Florian M Wurm 1*<br />

1 Laboratory of Cellular Biotechnology, Faculty of Life Sciences, Ecole<br />

Polytechnique Fédérale de Lausanne; 2 Laboratory of Hydraulic Machines,<br />

School of Engineering, Ecole Polytechnique Fédérale de Lausanne; 3 Chair of<br />

Modeling and Scientific Computing, School of Basic Sciences, Ecole<br />

Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland<br />

E-mail: florian.wurm@epfl.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P39<br />

Background: Orbitally shaken bioreactors (OSRs) of nominal volume from<br />

50 mL to 2’000 L have been developed for the cultivation of suspensionadapted<br />

mammalian cells. Here we study the hydrodynamics of OSRs for<br />

mammalian cells. The results are expected to allow the determination of<br />

key parameters for cell cultivation conditions and will facilitate the scaleup<br />

of OSRs.<br />

Materials and methods: CHO-DG44 cells were cultivated in suspension<br />

in 1-L bottles as described in [1]. To determine conditions under which<br />

the shear stress was harmful for the cells, the bottles were orbitally<br />

shaken on an ES-X platform (Kühner AG, Birsfelden, Switzerland) at<br />

agitation rates from 150 to 200 rpm for 24 h. Control cultures were run<br />

in parallel with agitation at 110 rpm. The velocity fields, shear stress,<br />

and free surface of a 1-L bottle at 110 rpm were simulated with<br />

Computational Fluid Dynamics (CFD). The Navier-Stokes equation was<br />

approximated with the finite element method. The simulations were all<br />

based on a mesh containing 50’000 tetrahedra and 10’000 vertices. The<br />

area of a finite element was 9.8 cm 2 . Because of the chosen<br />

discretization, each time step required the resolution of a linear system<br />

composed of 190’000 unknowns.<br />

Results: According to the CFD simulations of a 1-L bottle at 110 rpm, the<br />

maximal shear stress value was situated at the tip of the wave and was<br />

about 0.075 Pa (Figure 1a). The shear stress was higher at the walls than in<br />

the center of the liquid (Figure1b). Most of the zones in the liquid phase<br />

had a shear stress value equal to or lower than 0.02 Pa (Figure1b). The<br />

zones showing the maximal shear stress values represented less than 1%<br />

of the liquid phase (Figure1b).<br />

The cell growth rate was similar for the CHO culture agitated at 110 rpm<br />

and those at 150 or 160 rpm. According to CFD simulations, the maximal<br />

shear stress was ≤ 0.17 Pa at these higher agitation rates. At 170 rpm,<br />

the cell growth rate decreased and cell damage was observed. The<br />

maximal shear stress value at this agitation rate was about 0.19 Pa. The<br />

maximal values of shear stress were always situated at the tip of<br />

the wave independently of the agitation rate. The maximal value of shear<br />

stress increased with the agitation rate. However, only a small number of<br />

zones had these maximal values.<br />

Conclusions: The maximal shear stress value in a 1-L OSR agitated at<br />

110 rpm with a working volume of 300 mL was one to two orders of<br />

magnitude lower than values reported to be harmful for CHO cells [2]. This<br />

indicates that standard cultivation conditions in OSRs are safe for sensitive<br />

mammalian cells. The maximal shear stress was located at the tip of the<br />

wave of the free surface. However, as the wave of the free surface rotates<br />

with time, many zones of the liquid will be affected by the wave tip over<br />

time. Therefore, its potential to damage cells can not be neglected. Our<br />

study suggests that different wave patterns may lead to different maximal<br />

shear stress values. Further analysis of the correlation between the shape<br />

of the wave and the maximal shear stress level are required to determine a<br />

scale-up factor for hydrodynamic stress in OSRs.<br />

Acknowledgments<br />

We thank Dr. Mattia Matasci and Dr. Agata Oberbek for providing cell lines.<br />

We gratefully acknowledge Kühner AG and Sartorius-Stedim Biotech for the


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P39) The shear stress in a 1-L bottle at 110 rpm was simulated by CFD at the free surface (a) and in the liquid phase (cut view)<br />

(b). The shaking diameter was 5 cm and the working volume was 100 mL. The arrow indicates the tip of the wave.<br />

considerable support of equipment and material. This work was supported<br />

by the CTI Innovation Promotion Agency of the Swiss Federal Department of<br />

Economic Affairs and the Swiss National Science Foundation (SNSF).<br />

References<br />

1. Muller N, Girard P, Hacker DL, Jordan M, Wurm FM: Orbital shaker<br />

technology for the cultivation of mammalian cells in suspension.<br />

Biotechnol Bioeng 2005, 89:400-406.<br />

2. Tanzeglock T, Soos M, Stephanopoulos G, Morbidelli M: Induction of<br />

mammalian cell death by simple shear and extensional flows. Biotechnol<br />

Bioeng 2009, 104:360-370.<br />

P40<br />

A platform fed-batch process for various CEMAX® producer cell lines<br />

Benedikt Greulich * , Marika Poppe, Henry Woischnig, Karlheinz Landauer,<br />

Andreas Herrmann<br />

Celonic AG, Basel, Switzerland<br />

E-mail: Benedikt.Greulich@celonic.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P40<br />

The random nature of transgene integration harbours various pitfalls for<br />

development of production cell lines including clonal variation in<br />

expression level and growth characteristics. The CEMAX system is an<br />

expression system for targeted integration of expression cassettes via DNA<br />

double-strand break induced homologous recombination. Stable high<br />

producers are available within 4 weeks without the need of extensive<br />

clone screening and process development.<br />

Stable and high expression rates are ensured with the CEMAX system due<br />

to targeted integration of a single copy of the gene of interest at a<br />

transcriptionally highly active site in the host cell genome. Producer cell<br />

lines for various therapeutic product candidates were established. These<br />

cell lines produced antibodies and highly glycosylated antibody fusion<br />

proteins and showed high clonal similarity. This made a platform fed-batch<br />

process profitable.<br />

The CEMAX expression system is based on a genetically modified CHO cell<br />

line bearing a tag at a highly active genomic transcription site. The gene of<br />

interest (GOI) is integrated via site-directed DNA double-strand breakinduced<br />

homologous recombination. The target site comprises a screening<br />

and selection cassette, which was used for initial development of the high<br />

producer host cell. This exchangeable cassette is flanked by elements that<br />

facilitate site-directed integration by homologous recombination. These<br />

elements include regions of homology for recombination with the CEMAX<br />

Page 63 of 181<br />

vector, rudimentary selection markers that are activated during<br />

recombination, and cleavage sites for the homing endonuclease I-SceI<br />

(Meganuclease).<br />

Transfection of the CEMAX vector comprising the GOI along with transient<br />

Meganuclease activity triggers a chain of events after cotransfection: DNA<br />

gets cleaved by I-SceI and cellular DNA repair machinery is induced by free<br />

DNA double-strand breaks. The CEMAX vector comprising the GOI<br />

functions as a repair matrix using recombination between homologous<br />

elements flanking the DNA lesion. The GOI gets integrated and the<br />

rudimentary selection markers become activated upon homologous<br />

recombination. CEMAX producer cells were selected at multi-well plate<br />

scale followed by analysis for outgrowth of stable producer cells. The<br />

platform process for fed-batch cultivation of various CEMAX producer cells<br />

was applied after expansion to the scale needed for production of protein<br />

material.<br />

The idea of a platform process that is suitable for all CEMAX producer cells<br />

was based on theoretical consideration about similarity of cells due to sitedirected<br />

integration and the observation that cell growth and metabolism of<br />

CEMAX cell lines were comparable. This was verified by the results of this<br />

study. Applying the fed-batch process cell growth of CEMAX host cells and<br />

CEMAX producer cells was comparable (Figure 1). The development of the<br />

platform fed-batch process was based on three small scale development<br />

steps: (1) basal media screening, (2) feed medium optimization, and<br />

(3) improvement of feeding regime. Process development in 1 L bioreactors<br />

is currently ongoing.<br />

The fed-batch process comprises a chemically defined and commercially<br />

available basal medium and the chemically defined feed solution CeloFeed<br />

(Celonic) supplemented according to the improved feeding regime. The<br />

glucose level was maintained between 4.5 g/L and 7.5 g/L. Glutamine was<br />

kept at a concentration above 0.8 mM. By applying the platform fed-batch<br />

process product concentrations up to 690 mg/L were achieved with<br />

CEMAX cell lines producing a glycosylated Fc fusion protein after sitedirected<br />

integration of the GOI (Figure 1). Achievable productivity for<br />

different protein products varied from product to product. This was most<br />

probably due to different requirements in protein processing and<br />

secretion.<br />

The platform fed-batch process was developed by optimization based on<br />

commercial available and proprietary basal and feed media formulations. It<br />

was suitable for all producer cell lines derived of a particular CEMAX host<br />

cell due to highly similar growth behavior after site-directed integration of<br />

the gene of interest. The platform process made product concentrations of<br />

up to 0.7 g/L achievable. Animal derived component free chemically


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P40) Platform fed-batch process for CEMAX cell lines. Cells were cultivated in comparison to the CEMAX host cell line in shake flask<br />

scale. The basal medium was a commercially available chemically defined medium and was used in combination with the proprietary chemically defined<br />

feed solution CeloFeed. Product concentration was measured by Protein A HPLC.<br />

defined medium and feed solution ensure a robust and regulatory<br />

compliant process. By combining the platform process with site-directed<br />

integration timelines in cell line development and process development<br />

were shortened for faster entry in first-in-man studies.<br />

P41<br />

Development of a chemically defined CHO medium by engineering<br />

based on a feed solution<br />

Karlheinz Landauer * , Henry Woischnig, Natalie Hepp, Benedikt Greulich,<br />

Andreas Herrmann<br />

Celonic AG, Basel, Switzerland<br />

E-mail: Karlheinz.Landauer@celonic.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P41<br />

The use of chemically defined culture media for production of<br />

biopharmaceuticals has become state of the art. In combination with a<br />

suitable feed solution chemically defined media are the most potent driving<br />

factor for yield improvement in fed-batch processes. Although various<br />

chemically defined media are available of-the-shelf and deliver good results,<br />

those media are developed with the aim to sustain growth for various host<br />

cell lines, and need to be optimized for specific projects.<br />

We describe here the development of CeloCHO, a chemically defined<br />

medium for culture of recombinant Chinese hamster ovary cells. The<br />

medium was developed by engineering based on an optimised proprietary<br />

feed medium, which was developed previously. The approach was straightforward<br />

and linked superior results with minimum investments in medium<br />

development.<br />

In a first step of development, the components of the proprietary feed<br />

medium were categorized into five component groups: bulk salts, trace<br />

elements, vitamins, amino acids, and other components. Necessary<br />

components not present in the feed medium were considered while<br />

establishing a basic medium formulation based on theoretic considerations<br />

of cellular needs in culture (buffer system, adaptation of amino acid<br />

concentrations, glucose, growth factors, et cetera). This basic formulation<br />

was combined with various x-fold concentrates of the component group<br />

stock solutions.<br />

Seven media formulations were developed this way and were used for<br />

adaptation in the second step of development. At this stage cell densities<br />

up to 4×10 6 cells per ml in batch experiments were achieved using a CHO-<br />

K1 derived recombinant cell line producing a humanized antibody (IgG A).<br />

Albeit maximum viable cell density and integral of viable cell density were<br />

significantly less than achieved with the chemically defined reference<br />

Page 64 of 181<br />

medium for this cell line, product concentration was improved by 7% with<br />

the newly designed medium.<br />

The most suitable formulation was selected for fine tuning in the third phase<br />

of medium development. Therefore, the concentrations of the component<br />

groups were varied and individual components as well as the buffer system<br />

were optimized. Vitamins, for example, had a profound effect on cell growth<br />

and cell specific productivity. The integral of viable cells was reduced with<br />

vitamin concentrations, while productivity increased. Vitamin concentrations<br />

above 35% were not suitable for cultivation as observed by diminished cell<br />

growth.<br />

The final medium, CeloCHO, was tested with different CHO clones producing<br />

either IgG or fusion proteins. Figure 1 shows a batch experiment after<br />

adaptation of a recombinant cell line producing IgG A compared to the<br />

commercial available reference medium. Maximum viable cell density was<br />

improved 3-fold compared to the initial medium design with 12×10 6 cells<br />

per ml and exceeded the performance of the chemically defined reference<br />

medium. The product concentration was improved by 14% in the batch<br />

experiment and 57% in the fed-batch experiment compared to the<br />

reference medium (Table 1). Product concentration was 1.4 g/L without<br />

further process optimization. The improvement in fed-batch indicated a<br />

good compatibility of basal and feed medium. The feed medium that was<br />

used for supplementation was also used for the development of the initial<br />

basal medium formulation, which was probably the reason for the good<br />

performance. Product concentration in fed-batch experiments with other<br />

CHO-K1 derived producer cell lines was improved between 6% and 25%<br />

demonstrating suitability for different CHO-K1 derived cell lines (Table 1).<br />

The strategy for the development of a basal medium formulation based on<br />

a previously optimized feed solution proved suitable for medium<br />

development approaches without the need of extensive analytical work.<br />

Components were classified into five stock solutions based on chemical<br />

properties and combined as x-fold concentrates for fast media development.<br />

After a round of modifications regarding vitamins, buffer and salt<br />

optimization, cell densities up to 12×10 6 cells per ml were achieved in<br />

batch experiments with the chemically defined, animal derived component<br />

free CHO medium. The product concentration in fed-batch experiments<br />

was improved up to 57% compared to the reference medium for the cell<br />

line producing IgG A. Data of the third phase of medium development<br />

delivered a starting point for medium optimization to clonal demands,<br />

since the effects of several key components were studied in detail. Cell<br />

growth and specific productivity, for example, could be modulated in a<br />

vitamin-dependent way. Six cell lines were studied for growth and<br />

recombinant protein production using CeloCHO and demonstrated<br />

feasibility for universal use.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P41) Adaptation of cell line producing IgG A to CeloCHO and batch experiment. The experiment was performed in shake flask scale<br />

without process optimization.<br />

Table 1(abstract P41) Relative product concentration in batch and fed-batch mode using CeloCHO with 4 different cell<br />

lines<br />

Cell line, CHO-K1 derived IgG A, batch IgG A, fed-batch IgG B, fed-batch IgG C, fed-batch<br />

Reference medium a , Titer b<br />

100% 100% 100% 100%<br />

CeloCHO, Titer b<br />

114% 157% 125% 106%<br />

a b<br />

Chemically defined reference Medium optimized for each cell line. Normalized to product concentration in reference medium.<br />

P42<br />

Process development of ATROSAB, an anti TNFR1 Monoclonal<br />

Antibody: in three steps from research to GMP<br />

Karlheinz Landauer 1* , Cuneyt Unutmaz 1 , Simon Egli 1 , Verena Berger 2 ,<br />

Simone Lais 1 , Timo Liebig 1 , Daniel Steiner 1 , Jo Maier 1 , Ina Rostalski 1 ,<br />

Francois Forcellino 1 , Andreas Herrmann 1,2<br />

1 Celonic AG, Basel, Switzerland; 2 Celonic GmbH, Jülich, Deutschland<br />

E-mail: Karlheinz.Landauer@celonic.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P42<br />

The humanized monoclonal antibody ATROSAB is targeting specifically<br />

the TNF receptor 1(TNFR1). TNF is a central mediator of inflammation and<br />

key target for intervention in inflammatory diseases such as rheumatoid<br />

arthritis, psoriasis and Crohn’s Diseases. Notably, blockade of the second<br />

TNF receptor, TNFR2, has been associated with increased sensitivity to<br />

viral infections or increased susceptibility to demyelinating disorders and<br />

lymphomas. In this context, a selective inhibition of TNF-induced TNFR1<br />

but not TNFR2 signaling activity holds great promises to overcome<br />

undesired effects observed with less specific TNF antagonists currently<br />

used in clinic.<br />

The recently humanized antibody was used to establish a rCHO-K1 cell line<br />

under serum-free, chemically defined conditions (1). The process<br />

development was based on two sets of shake flask experiments, three 10L<br />

bioreactor runs and finally 2 GMP production runs in 300 L scale. The scale<br />

up strategy was based on quality by design considerations specified in the<br />

design space for the scale-up parameters. The main parameter for the scaleup<br />

development was mixing time as a function of the bioreactor geometry,<br />

tip-speed, Reynolds number, and the power input of the systems. Those<br />

parameters are generally considered as important parameters during upstream<br />

process development.<br />

Protein characteristics were specified based on several considerations.<br />

A major part was the ICH-Q8 guideline as well as antibody and other<br />

Page 65 of 181<br />

drug guidelines. The other part is based on experience with antibody<br />

development and the specific protein in special. The specifications are<br />

described in Table 1. The analytical methods used to characterize the<br />

antibody were SDS-PAGE, western blot, HPLC-SEC, protein A HPLC,<br />

HPAEC-PAD, Isoelectric focusing and a cell based potency assay. As the<br />

antibody is declined to bind to the TNF-R1 on target cells, it was<br />

engineered not to elicit ADCC or CDC. Therefore those parameters were<br />

not checked during process development.<br />

The basal medium was fixed to a commercial available chemically defined<br />

medium during clone screening. In a batch process, performed in a fully<br />

equipped 1L bioreactor, a titer of approximately 180 mg/L was obtained.<br />

At this stage the antibody was characterized and specifications were<br />

partly narrowed and some were newly set. In the first step of process<br />

development different feeding solutions and concentrations thereof were<br />

tested in shake flask experiments. Metabolite data were monitored on a<br />

daily basis comprising glucose, lactate, ammonia, and amino acid levels.<br />

Additionally cell density and protein concentration were measured at the<br />

same time interval. For in-depth protein characterization, supernatant was<br />

purified after 6 days of fermentation and at harvest, in order to check the<br />

quality attributes already early at the development stage. This step led to<br />

an increase of about 2.5 fold in product concentration, while the protein<br />

characteristics stayed within specifications at both sampling time points.<br />

The second step was the optimization of the feeding strategy as well as<br />

the fortification of the solutions with certain chemically defined<br />

supplements. This led to another increase of about 2.5 fold regarding<br />

final product concentration. Product quality changed slightly, but stayed<br />

within specifications. Especially the glycosylation structure changed<br />

towards less glycosylated versions. However, no impact on the bioassay<br />

was observable. The process was finally scaled up to the 300L stirred tank<br />

bioreactor in the GMP facility. An engineering run was performed to test<br />

the process and product parameters. Some influence was measured, thus<br />

the process was slightly changed prior the first GMP run. The obtained<br />

product quality was well within specifications, the achieved titer was


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P42) specified scale-up parameters and defined protein characteristics<br />

scale-up parameter protein characteristics<br />

Parameter Design Space 300L GMP specifications defined at the<br />

project start<br />

300L GMP<br />

Mixing Time [s] 10 - 70 max. 60 Isoforms (IEF) pH 7.6 - 8.8<br />

Tip-Speed [m/s] 0.6 - 1.2 max. 1.0 Glycosylation<br />

Mol% of different forms ± 20% from clone<br />

(HPAEC-PAD)<br />

screening<br />

Reynolds<br />

Number<br />

1 - 8 Mio max. 6 Mio Potency 40% - 160% from clone screening<br />

Power Input [W/ 25 - 1100 max. 650 Molecular weight<br />

Main Band at app. 150 kDa<br />

m3]<br />

(SDS-PAGE)<br />

Gassing Head Space / Ring Head Space & Ring Immunogenic glycostructures none<br />

Sparger<br />

Sparger<br />

Agitation orbital sh., Rushton I. Rushton I. Purity (HP-SEC) > 90%<br />

Figure 1(abstract P42) The left diagram shows the cell density of two GMP runs in the 300L bioreactor. The right diagram focuses on the achieved<br />

product concentrations during the different process development steps. The batch experiment was performed in 1L bioreactors, the feed screening was<br />

done in 250 mL shake flasks. The feed optimization was performed in 10L bioreactors and the GMP runs were done in a 300L stirred tank bioreactor in a<br />

GMP facility.<br />

more than 1.2 g/L, which was a more than 7 fold increase in comparison<br />

to the initial batch process. Although the cell density profile of the<br />

process changed between run 1 and 2, the volumetric product concentration<br />

was only slightly below 1.2 g/L and the product characteristics<br />

were identical to the first run. This indicates the robustness of the<br />

developed process and the validity to define and specify design spaces<br />

for process development especially during scale-up development. The<br />

analysis of protein characteristics employing SDS-PAGE, IEF, HP-SEC,<br />

potency assay and glycosylation pattern with HPAEC-PAD ensures the<br />

quality of the protein.<br />

Reference<br />

1. Zettlitz KA, Lorenz V, Landauer K, Münkel S, Herrmann A, Scheurich P,<br />

Pfizenmaier K, Kontermann R: ATROSAB, a humanized antagonistic antitumor<br />

necrosis factor receptor one-specific antibody. mAbs 2 2010,<br />

2(6):639-647.<br />

P43<br />

Cell line development using the SEFEX system<br />

Benedikt Greulich 1* , Verena Berger 2 , Marika Poppe 1 , Natalie Hepp 1 ,<br />

Karlheinz Landauer 1 , Andreas Herrmann 1,2<br />

1 Celonic AG, Basel, Switzerland; 2 Celonic GmbH, Jülich, Deutschland<br />

E-mail: Benedikt.Greulich@celonic.ch<br />

BMC Proceedings 2011, 5(Suppl 8):P43<br />

Page 66 of 181<br />

Cell lines producing biopharmaceuticals with high yield and high quality<br />

in a regulatory compliant environment are a prerequisite for cost<br />

effective bioproductions. The development of these production cell lines<br />

often includes screening strategies combined with gene amplification and<br />

limited dilution experiments, a time consuming process. Especially gene<br />

amplification tends to interfere with clonal stability. Limited dilution,


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

especially using a serum-free culture environment is prone to failure and<br />

low clone yields.<br />

We present here the SEFEX platform technology for the development of<br />

non-amplified high yield production cell lines. The strategy is based on a<br />

regulatory compliant method for transfection and single cell cloning using<br />

a proprietary, fully tested, CHO-K1 host cell line adapted to chemically<br />

defined medium. Fast track cell lines were generated by selection of single<br />

cell derived clones within 2.5 months after transfection. 1.0 g/L product<br />

concentration were achieved after two rounds of process development<br />

using these cell lines. Optimized cell lines were developed based on fast<br />

track cell lines employing a second transfection. These cell lines were<br />

capable for production of 2.6 g/L during an early phase of process<br />

development.<br />

Cell line development with the SEFEX technology comprises steps carried<br />

out entirely under serum-free conditions including transfection, selection,<br />

and single cell cloning. Fast track cell lines were developed within 2.5<br />

months by transfection and selection of stably transfected cells followed by<br />

single cell cloning. During single cell cloning, semi-automated photo<br />

documentation at the single cell level is used to assure and document<br />

clonality in a regulatory compliant manner. Table 1 summarizes cell specific<br />

productivity and product concentration in fed-batch processes. More than<br />

1.0 g/L were achieved in a GMP process at 300 L scale after two steps of<br />

process development followed by scale-up development. A similar scale-up<br />

strategy including GMP production was summarized by Landauer et al. in<br />

Page 67 of 181<br />

this issue [1]. Nevertheless, production of proteins that are difficult to<br />

express like, IgG C in Table 1, suffered from low productivity and low yield.<br />

Optimized cell lines, which were obtained by a second serial transfection of<br />

the antibody expression vector including regulatory sequences, were<br />

suitable to deliver high rates of antibody production. The productivity of IgG<br />

C was improved 6-fold to18 pg/c/d. This allowed production of the difficult<br />

to express protein at a product concentration of 1.1 g/L in fed-batch mode<br />

(Table 1). Other examples of optimized cell lines provided product<br />

concentrations of 1.7 g/L without any optimization during cell line<br />

development (IgG A, Table 1), or 2.6 g/L after two steps of upstream process<br />

development at 1 L bioreactor scale (IgG B, Table 1, Figure 1).<br />

Figure 1 shows growth curves of the fast-track and optimized cell line<br />

producing IgG B. Optimization of maximum viable cell density was not<br />

addressed during upstream process development so far. This optimization<br />

is currently ongoing and leaves room for further improvements. Specific<br />

productivity of the optimized cell lines varied between 18 and 36 pg/c/d,<br />

which was equivalent to a two- to six-fold improvement compared to the<br />

fast track cell lines. Productivity was suitable for production of high volume<br />

products. Scale-up to 300 L stirred tank and 1000 L wave bioreactor for<br />

production of clinical phase II drug product was shown for production cell<br />

lines generated with the SEFEX platform technology.<br />

The SEFEX platform technology for the development of non-amplified high<br />

yield production cell lines is based on a regulatory compliant, proprietary<br />

CHO-K1 host cell line adapted to chemically defined medium. Fast track<br />

Table 1(abstract P43) Cell specific productivity and product concentration obtained with fast track cell lines and<br />

optimized cell lines<br />

specific productivity [pg/cell/day] product concentration fed-batch [g/L]<br />

Project fast track cell line optimised cell line fast track cell line optimised cell line<br />

1 (IgG A) 11 36 0.6 a<br />

1.7 a<br />

2 (IgG B) 12 25 1.0 b<br />

2.6 c<br />

3 (IgG C) 3 18 no fed-batch 1.1 d<br />

a b<br />

Fed-batch experiment during cell line development without any optimization using chemically defined basal medium and proprietary CeloFeed. Fed-batch<br />

experiment after two steps of process development and scale-up to 10 L using chemically defined basal and feed medium. c Fed-batch experiment at 1 L<br />

bioreactor scale after two steps of optimization using chemically defined basal medium and proprietary CeloFeed. d Fed-batch experiment at shake flask scale<br />

using chemically defined basal medium and proprietary CeloFeed.<br />

Figure 1(abstract P43) Fed-batch experiments for production of IgG B. The fast-track cell line (left hand graph) was cultivated at 10 L scale after two<br />

steps of process development and scale up using chemically defined basal and feed medium. The optimized cell line (right hand graph) was cultivated<br />

at 1 L scale after basal media and feed media screening. A chemically defined medium was used in combination with the proprietary feed solution<br />

CeloFeed.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

cell lines, which were available within 2.5 months produced 1.0 g/L<br />

product. Optimized cell lines, which were developed based on fast track<br />

cell lines, were capable for production of 2.6 g/L in early phase of process<br />

development. Using the SEFEX platform technology, all development steps<br />

were carried out under entirely serum-free or chemically defined media<br />

conditions including transfection, selection, and single cell cloning.<br />

Clonality was assured and documented in a regulatory compliant manner<br />

using photo documentation of single cells in cell cloning experiments. The<br />

serial transfection cell line development strategy described here provides<br />

the possibility to develop production cell lines meeting industrial demands<br />

employing simplest process development procedures within minimized<br />

time frames.<br />

Reference<br />

1. Landauer K, Unutmaz C, Egli S, Berger V, Lais S, Liebig T, Steiner D, Maier J,<br />

Rostalski I, Forcellino F, Herrmann A: Process Development of ATROSAB,<br />

an anti TNFR1 Monoclonal Antibody: in Three Steps from Research to<br />

GMP. BMC Proceedings 2011 in press, this issue.<br />

P44<br />

Platform process for production of monoclonal antibodies for research<br />

purposes – improvement option<br />

Joern Meidahl Petersen 1* , Claus Kristensen 2<br />

1 Biopharm Manufacturing Development, API Support, Gentofte, Denmark,<br />

DK-2820; 2 Biopharmaceutical Research, Mammalian Cell Technology,<br />

Maaloev, Denmark, DK-2760<br />

E-mail: jmp@novonordisk.com<br />

BMC Proceedings 2011, 5(Suppl 8):P44<br />

Background: In 2006 Novo Nordisk decided to invest in building a pipeline<br />

within inflammatory disease management. To support this strategy the cell<br />

culture units had to establish technology for expression and production of<br />

Figure 1(abstract P44) Yields of 11 anitbodies in shake flask and bioreactors.<br />

monoclonal antibodies in CHO cells. A number of technology providers at<br />

that time offered proven platform processes for this purpose. It was decided<br />

to in-license one of these technology platforms, the one developed at Lonza<br />

Biologics, and focus research resources on product innovation rather than<br />

development of an in-house production system. The platform has now been<br />

fully implemented and the work flow optimised and standardised.<br />

Platform process review: The platform process comprises:<br />

Host cell line/expression system<br />

Medium/feeds (chemically defined – animal derived component free)<br />

Process parameters<br />

Scale-down model (shake flask, 100 ml working volume).<br />

The platform process has been applied for cell line development and<br />

antibody production for R&D purposes for five years. During this period 11<br />

monoclonal antibodies have been transferred from laboratory scale to pilot<br />

plant production. The performance of the process platform across projects<br />

has been reviewed. The scope of the review was to compare yields obtained<br />

in the scale down model and yields obtained in the bioreactor process for<br />

all 11 antibodies. Figure 1 shows the results of the review.<br />

In conclusion the review show that<br />

The cell lines are yielding 1.9 – 4.5 g/l mAb.<br />

The scale down model is predictive for the bioreactor process.<br />

Evaluation of an improvement option: An upgrade of the medium,<br />

feeds and process protocols was offered by Lonza Biologics. A b-version<br />

of the latest process from Lonza has been tested and compared to the<br />

previous version in a study including five cell lines. The experiments were<br />

carried out in 100 ml shaker flask and the scale down version of the<br />

process was applied. A comparison of the two fed batch procedures is<br />

shown below:<br />

Current version: Version 6<br />

Medium and feeds CD-ACF<br />

Two feed solution<br />

Feed rates based on<br />

Table 1(abstract P44) Result of the comparison of the two versions of the fed batch process<br />

mAb Current version: Version 6 New version: Version 8<br />

ICA* )<br />

Lactate mAb ICA Lactate mAb<br />

10E6 viable cell days/ml mmol/l g/l 10E6 viable cell days/ml mmol/l g/l<br />

C 108 57 3.5 92 28 7.1<br />

D 103 61 2.2 90 29 5.2<br />

E 162 65 5.5 104 17 9.0<br />

H 100 55 3.2 120 25 7.0<br />

J 148 62 3.2 164 21 4.3<br />

* ) ICA: Integrated Cell Area.<br />

Page 68 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

○ Viable Cell Density<br />

○ Residual glucose<br />

New version: Version 8<br />

Medium and feeds CD-ACF<br />

Five feed solutions<br />

Feed rates based on<br />

○ Viable Cell Density<br />

○ Residual glucose<br />

○ Time<br />

The results of the study are compiled in table 1.<br />

The comparison of the b-test of improved platform process and the<br />

current process showed:<br />

mAb yields improved 1.3 – 2.4 fold<br />

No change in integrated cell area<br />

Improved lactate control may be the key to the yield improvement.<br />

Acknowledgements: The following people all contributed with cell lines,<br />

data and/or stimulating discussions<br />

Novo Nordisk:Birgitte Friedrichsen, Bjarne Rask Poulsen, Mark Chadfield,<br />

Ann Merete Mygind, Ida Mølgaard Kundsen, Max Wellerdiek, Gitte<br />

Johnsen.<br />

Lonza Biologics:Alison Porter, Jeetendra Vaghjiani.<br />

Figure 1(abstract P45) Cluster analysis of MALDI-TOF MS spectra from 92 different cell lines.<br />

Page 69 of 181<br />

P45<br />

Development and validation of a protocol for cell line identification by<br />

MALDI-TOF MS<br />

Guido Vogel 1* , André Strauss 1 , Bernard Jenni 1 , Dominik Ziegler 1 ,<br />

Eric Dumermuth 2 , Sylvie Antz 2 , Claudia Bardouille 3 , Beat Wipf 3 ,<br />

Christian Miscenic 3 , Georg Schmid 3 , Valentin Pflüger 1<br />

1 Mabritec AG, 4125 Riehen, Switzerland; 2 Novartis Pharma AG , 4056 Basel,<br />

Switzerland; 3 F. Hoffmann-La Roche AG, 4070 Basel, Switzerland<br />

E-mail: guido.vogel@mabritec.com<br />

BMC Proceedings 2011, 5(Suppl 8):P45<br />

Summary: Misidentification or cross-contamination of cell lines used in<br />

biotechnology or diagnostic settings are a challenge for laboratories and cell<br />

culture repositories. Masters et al. [1] among others reported the occurrence<br />

of large numbers of unrecognized and unreported misidentification or crosscontamination<br />

of cell lines. Current methods for the authentication of cell<br />

lines such as karyotyping, 2D-gel-electrophoresis, restriction fragment length<br />

polymorphism (RFLP) or short tandem repeats (STR) are expensive, labor<br />

intensive and not routinely applied. In the last decade MALDI-TOF MS<br />

became a powerful tool for the rapid and cost effective identification and


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P45) Example of problematic samples<br />

sample name expected species MALDI-TOF MS results<br />

Sf9 Spodoptera frugiperda Trichoplusia ni<br />

HepG2 Homo sapiens Mus musculus<br />

SK-MEL-30 Homo sapiens Homo sapiens profile with strongly altered mass composition: contamination possible<br />

CHO-K1 Cricetulus griseus unknown mass profile<br />

J558L Mus musculus Mus musculus profile with strongly altered mass composition: contamination possible<br />

WS1 Homo sapiens unknown mass profile<br />

taxonomic classification of microorganisms directly from whole cell extracts.<br />

We adapted this protein fingerprinting approach for a fast species<br />

identification of eukaryotic cell lines and established as well as validated a<br />

reference database.<br />

In addition we demonstrate that this new approach has a potential for the<br />

rapid characterization of recombinant protein expression systems. Accurate<br />

protein expression and the determination of the molecular mass of the<br />

recombinant expressed proteins (20-120kDa) can directly be analyzed from<br />

stable transfected and virus infected cultures.<br />

Materials and methods: 1 ml of fresh culture was transferred into an<br />

Eppendorf tube and washed once with 1x PBS. The pellet was resuspended<br />

in 70% EtOH for transport and storage at RT. 1µl of the cell pellet was<br />

transferred with a plastic loop to a new Eppendorf tube and mixed with<br />

20µl formic acid (10%). The suspension was mixed with 40µl of saturated<br />

sinapinic acid matrix. 1µl of sample suspension was spotted on a steel target<br />

plate in duplicates. Spectra were acquired on Shimadzu Confidence MALDI-<br />

TOFMSinlinearmodeinamassrangefrom2-50kDa.Peaklistswere<br />

imported into SARAMIS software for Marker pattern definition and<br />

automated identification.<br />

Results: In a first phase we analyzed 146 different eukaryotic cell lines<br />

derived from 11 different taxa (Figure 1), including common lines from<br />

culture collections as well as customer specific in-house lines. Marker<br />

patterns were established for these 11 taxa and validated in a blinded<br />

study with 9 mammalian and 48 insect cell lines. All 57 test samples were<br />

correctly identified on the species level. Even two intentionally mixed<br />

samples were assigned to the right species.<br />

In a second phase we analyzed more than 178 mammalian and 240<br />

insect cell lines for identity authentication from different customers in<br />

order to evaluate the usefulness of this service. Out of 418 samples<br />

analyzed, we revealed 6 samples with either wrong identities, strongly<br />

altered or unknown mass profiles (Table 1).<br />

When necessary cell lines were also analyzed for their expression profile<br />

after infection or transfection with viral vectors. It was possible to<br />

determine precise masses of overexpressed proteins as compared to<br />

controlled samples. For these additional experiments, no further sample<br />

preparation was necessary.<br />

Conclusion: We developed a fast, simple and accurate method with high<br />

throughput capacity for the authentication of cell lines. Since the<br />

construction of customer specific mass profile libraries is straightforward,<br />

we propose this approach as an additional method for routine cell line<br />

authentication in biotechnology settings.<br />

Further we demonstrated that MALDI-TOF MS is also a fast and simple<br />

tool for the characterization of eukaryotic protein expression systems,<br />

especially for the determination of a precise mass.<br />

Reference<br />

1. Masters JR, et al: Short tandem repeat profiling provides an international<br />

reference standard for human cell lines. PNAS 2001, 98:8012-8017.<br />

P46<br />

DoE of fed-batch processes – model-based design and experimental<br />

evaluation<br />

Onur Sercinoglu 1 , Oscar Platas Barradas 1 , Volker Sandig 2 , An-Ping Zeng 1 ,<br />

Ralf Pörtner 1*<br />

1 Institute of Bioprocess and Biosystems Engineering, Hamburg University of<br />

Technology, Hamburg, D-21073, Germany; 2 ProBioGen AG, Berlin, D-13086,<br />

Germany<br />

E-mail: poertner@tuhh.de<br />

BMC Proceedings 2011, 5(Suppl 8):P46<br />

Page 70 of 181<br />

Background: Experimental process-development and optimization is<br />

expensive and time-consuming. Real optimization by means of design of<br />

experiments involves data generation before optimization can be aimed<br />

for. This can make the way from process development to process<br />

establishment even harder, since academia or start-up research facilities<br />

might not have the possibility to generate these data. Furthermore,<br />

bioprocesses involving mammalian cells deal with many critical variables;<br />

processes are not only carried out batch wise, but increasingly in fedbatch<br />

mode with desired feeding profiles.<br />

The use of DoE tools in combination with an appropriate growth model<br />

might allow the experimenter to develop and to test fed-batch strategies<br />

in silico, before experiments are carried out in the laboratory.<br />

In our work, an unstructured model for mammalian cell culture was used<br />

for simulation. Kinetic parameters were derived from a small number of<br />

shake-flask experiments. The model was tested for data generation on<br />

common fed-batch strategies. By means of design of experiments<br />

strategies, relevant conditions were selected and experimentally tested. In<br />

this way, suitable fed-batch strategies for mammalian cell lines are<br />

evaluated in silico before bioreactor experiments are to be performed.<br />

This results in a significant reduction in the number of experiments<br />

during process development for mammalian cell culture.<br />

Concept – fed-batch Analyzer -: A tool was developed in Mathworks‘<br />

Matlab for simulation of batch and fed-batch cultivations of mammalian<br />

cells. This program was created as a GUI (Guided User Interface), in which<br />

the user will be guided through the process of developing a fed-batch<br />

strategy. Growth models can be selected as well as different fed-batch<br />

modes. For a manageable comparison of fed-batch strategies, in silico<br />

experiments can be planned by design of experiments (DoE) and initial<br />

conditions as well as cell-line-specific kinetic parameters can be obtained<br />

from a reduced number of small-scale experiments e.g. shake flasks. The<br />

following steps will explain the procedure for in silico experimentation<br />

during the development of a fed-batch strategy for the human<br />

production cell line AGE1.hn (ProBioGen AG). Afterwards, one process<br />

condition will be chosen and tested in the laboratory.<br />

Step 1: Define the process model: Growth models [1,2] have been<br />

predefined for operation of the tool. However, the user can modify or<br />

include new models according to her/his expertise and knowledge of the<br />

cell line. For determination of model parameters, a reduced number of<br />

experiments in small scale (e.g. shake flasks), is to be performed.<br />

Step 2: Load the model: Step 3: Choose a feeding strategy: Simple<br />

feeding strategies have been predefined in order to make the transfer of<br />

in silico results into laboratory experimentation easier. Constant feed,<br />

linear feed, exponential feed and step feed can be selected. New feeding<br />

profiles can be added to the program to improve its possibilities.<br />

Step 4: Create design: The design (e.g. full factorial design) can be<br />

either planed within the program (MATLAB Statistics Toolbox) or loaded<br />

from other DoE tools (e.g. Design Expert). In order to define the central<br />

point for the design, data from batch experiments can be used.<br />

Step 5: Run design: The tool conduct the in silico experiments and<br />

delivers a response. This is normally a variable the user is interested in<br />

(IVCD, VCD or product concentration).<br />

Step 6: Obtain in silico results: For each set of conditions, the tool<br />

generates a sheet with virtual curves. For an overview of results and<br />

better comparison of the process conditions, a response curve can be<br />

generated (see Figure 1).<br />

Step 7: Implementation in the laboratory: Following experiment was<br />

planned and performed in a 1 L Bioreactor. (Table 1, Figure 2)<br />

Acknowledgements: Funding by the BMBF, Grand Nr. 0315275A is<br />

gratefully acknowledged.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P46) Surface response plan for a feeding strategy with constant feed rate.<br />

References<br />

1. Jang JD, Barford J.P: An Unstructured Kinetic Model of Macromolecular<br />

Metabolism in Batch and Fed-Batch Cultures of Hybridoma Cells<br />

Producing Monoclonal Antibody. Biochemical Engineering Journal<br />

4(2):153-168.<br />

2. Zeng A-P, Deckwer W-D: Mathematical modeling and analysis of glucose<br />

and glutamine utilization and regulation in cultures of continuous<br />

mammalian cells. Biotechnology and Bioengineering 1995, 47:3.<br />

Table 1(abstract P46) Experimental conditions for<br />

validation of a contant feeding strategy<br />

Parameter Value<br />

Initial conditions (batch)<br />

Glucose 10 mM<br />

Glutamine 2 mM<br />

fed-batch strategy<br />

Mode constant<br />

Glucose in Feed 60 mM<br />

Glutamin in Feed 8 mM<br />

Feed Rate 0.059 mL min -1<br />

Feed Start 24 h<br />

Page 71 of 181<br />

Figure 2(abstract P46) Experimental results after culture of AGE1.HN<br />

cells in bioreactor with a constant feed rate.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

P47<br />

Criteria for bioreactor comparison and operation standardisation<br />

during process development for mammalian cell culture<br />

Oscar Platas Barradas 1 , Uwe Jandt 1 , Linh Da Minh Phan 1 , Mario Villanueva 1 ,<br />

Alexander Rath 2 , Udo Reichl 2 , Eva Schräder 3 , Sebastian Scholz 3 , Thomas Noll 3 ,<br />

Volker Sandig 4 , Ralf Pörtner 1* , An-Ping Zeng 1<br />

1 Institute of Bioprocess and Biosystems Engineering, Hamburg University of<br />

Technology, Hamburg, D-21073, Germany; 2 Bioprocess Engineering, Max<br />

Planck Institute for Dynamics of Complex Technical Systems, Magdeburg,<br />

D-39106, Germany; 3 Faculty of Technology, AG Zellkulturtechnik, Bielefeld<br />

University, Bielefeld, D-33501, Germany; 4 ProBioGen AG, Berlin, D-13086,<br />

Germany<br />

E-mail: poertner@tuhh.de<br />

BMC Proceedings 2011, 5(Suppl 8):P47<br />

Background: Development of bioprocesses for animal cells has to deal with<br />

different bioreactor types and scales. Bioreactors might be intended for<br />

generation of cell inoculum and production, research, process development,<br />

validation or transfer purposes. During these activities, not only the difficulty<br />

of up- and downscaling might lead to failure of consistency in cell growth,<br />

but also the use of different bioreactor geometries and operation<br />

conditions. In such cases, the criteria for bioreactor design and process<br />

transfer should be carefully evaluated in order to avoid an erroneous<br />

transfer of cultivation parameters.<br />

Page 72 of 181<br />

In this work, power input, mixing time, impeller tip speed, and Reynolds<br />

number have been compared systematically for the cultivation of the<br />

human cell line AGE1.HN® within three partner laboratories using five<br />

differentbioreactorsystems.Acommonprocesswindowformixing<br />

time in the range of 8 – 13 s has been found in bioreactors having<br />

significant differences in their inner geometries. The obtained results<br />

are employed for process standardisation and transfer between<br />

research institutions.<br />

Cell culture in laboratory bioreactors with different inner geometries:<br />

Finding conditions for consistent cultivation of mammalian cells in<br />

bioreactors is not an easy task. For standard stirred tanks, correlations<br />

existing in literature can be used in order to predict operation conditions for<br />

process transfer purposes. However, if the inner geometry of two bioreactors<br />

cannot be compared within tolerance ranges, the characterization of the<br />

bioreactor hydrodynamics becomes necessary.<br />

For this work, five geometrically different bioreactors were used, which<br />

are operated within three partner laboratories for data generation<br />

during research on Systems Biology. Characterization of the bioreactor<br />

hydro-dynamics was performed with the main goal of finding a<br />

relationship between process transfer criteria and cell growth in the<br />

systems.<br />

Bioreactor characterization: Following criteria were considered for<br />

characterization of bioreactor hydrodynamics.<br />

(1) Power input (P/V): Power numbers Np were calculated from Np = f<br />

(Re) correlations available in literature [1-3]. Corrections for Np were<br />

Figure 1(abstract P47) Relationship between maximum specific growth rate μ max and values for process transfer during bioreactor culture:<br />

a) Power input, b) Mixing time, c) Impeller tip speed, and d) Reynolds number. Curve fitting for bioreactors 3 (Black square) and 5 (Purple circle).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

considered due to geometry deviations from a standard configuration<br />

[4-7]. Volumetric power inputs were calculated according to Equation 1:<br />

P<br />

V<br />

=<br />

NpN d<br />

V<br />

3 5 r<br />

(2) Mixing time (Θ 94.5): This criterion was obtained from the<br />

decolourization of a I/KI solution of after addition of Na 2S 2O 3.Starchwas<br />

added previously to the bioreactor. Decolourization time course was<br />

video recorded. The resulting videos were computer analyzed, and Θ 94.5<br />

was obtained after gray-scale conversion and measurement of loss of<br />

saturation (MATLAB, MathWorks).<br />

(3) Impeller tip speed (u tip): calculated according to Equation 2.<br />

utip = p Ndi<br />

(2)<br />

(4) Reynolds Number at impeller tip (Re i): calculated according to<br />

equation 3:<br />

Re i<br />

Ndi<br />

= r<br />

2<br />

h<br />

The specific growth rate μmax was employed as indicator for comparison<br />

of bioreactor performance. The use of μmax made the comparison of the<br />

two cell line clones possible, despite the differences in initial cell<br />

densities during bioreactor culture.<br />

Relationship between cell growth and process transfer criteria:<br />

Figure 1 shows the dependency of the μmax on process transfer criteria. A<br />

process window for mixing time values between 8 and 13 seconds can<br />

be identified as common for all bioreactors, where the smallest deviation<br />

in µ max between different bioreactors can be observed.<br />

Conclusions: Criteria for process transfer were analyzed during the<br />

cultivation the human production cell line AGE1.HN. Growth was<br />

compared within ranges for power input, mixing time, impeller tip speed<br />

and Reynolds number. Maximum specific growth rates were observed for<br />

AGE1.HN cells at a common mixing time range of 8 - 13 seconds for all<br />

cultivation systems. This criterion was observed to be a reference for<br />

consistency of results within laboratory bioreactors with different internal<br />

geometry.<br />

Funding by the BMBF, Grand Nr. 0315275A is gratefully acknowledged.<br />

Nomenclature:<br />

di impeller diameter [m]<br />

M degree of mixing<br />

N agitation speed [rpm]<br />

P = NprN 3 5<br />

di power input [W m -3 ]<br />

Rei Reynolds number at impeller tip [-]<br />

utip impeller tip speed [m s -1 ]<br />

V working volume [m 3 ]<br />

Greek letters:<br />

Θ mixing time [s]<br />

μmax specific growth rate [d -1 ]<br />

r density [kg m -3 ]<br />

h dynamic viscosity [kg m -1 s -1 ]<br />

References<br />

1. Rushton JH: Power Characteristics of Mixing Impellers. Part I. Chem. Eng.<br />

Prog 1950, 46(8):395-404.<br />

2. Rushton JH: Power Characteristics of Mixing Impellers. Part II. Chem. Eng.<br />

Prog 1950, 46(9):467-476.<br />

3. Bates RL: An examination of some geometric parameters of impeller<br />

power. I&EC Process Design and Development 1963, 2(4).<br />

4. Markopoulos J, Pantuflas E: Power consumption in gas-liquid contactors<br />

agitated by double-stage rushton turbines. Chem. Eng. Technol 2001,<br />

24(11):1147-1150.<br />

5. Henzler H-J: Verfahrenstechnische Auslegungsunterlagen fur<br />

Rührbehalter als Fermenter. Chem.-Ing.-Tech 1982, 54(5):461-476.<br />

6. Hudcova V, Machon V, Nienow AW: Gas-Liquid dispersion with dual rushton<br />

turbine impellers. Biotechnology and Bioengineering 1988, 34:617-628.<br />

7. Hemrajani RR: Mechanically Stirred Vessels. Handbook of Industrial Mixing.<br />

Science and Practice Wiley-Interscience 2004.<br />

(1)<br />

(3)<br />

Page 73 of 181<br />

P48<br />

Cultivation strategies of a BA/F3 cell line for fundamental cell<br />

research<br />

Martin Schaletzky 1 , Oscar Platas Barradas 1 , Henning Sievert 2 ,<br />

Stefan Balabanov 2 , An-Ping Zeng 1 , Ralf Pörtner 1*<br />

1 Institute of Bioprocess and Biosystems Engineering, Hamburg University of<br />

Technology, Hamburg, D-21073, Germany; 2 Cancer Center Hamburg,<br />

University Medical Center Hamburg-Eppendorf (UKE), Hamburg, D-20246,<br />

Germany<br />

E-mail: poertner@tuhh.de<br />

BMC Proceedings 2011, 5(Suppl 8):P48<br />

Background: During the chronic myelogenous leukemia (CML) the Bcr-Abl<br />

oncoprotein is produced, which leads to unregulated cell proliferation.<br />

CML is treated with one of several targeting therapies such as imatinib,<br />

(formerly STI-571 [Glivec; Novartis, Switzerland]) a selective inhibitor that<br />

blocks tyrosin kinase activity of the Bcr-Abl oncoprotein. Apart from the<br />

second generation Bcr-Abl inhibitors, identifying novel direct or indirect<br />

downstream targets of Bcr-Abl could contribute significantly to the<br />

development of new synergistic treatment strategies against CML. The<br />

effects of imatinib on the protein expression of Bcr-Abl positive cells are<br />

being investigated [1]. A protein which is downregulated during treatment<br />

with imatinib (eukaryotic translation initiation factor eIF5A) was identified.<br />

This protein is a potentially promising target for single-agent and<br />

combined-treatment strategies for CML. For protein complex identification<br />

a high cell number is needed. This is difficult to be obtained reproducibly<br />

withflaskculturesorrollerbottles.Theaimofthisprojectwastodevelop<br />

and establish a reproducible bioreactor cultivation of murine suspension<br />

cell lines (BA/F3 p210), which yields a total cell number close to 1·10 10 cells<br />

required for analytics. Cells should be in exponential growth under<br />

constant culture conditions at the time of harvest. A small stirred tank<br />

bioreactor with a working volume of 150 mL was used to study and<br />

compare different operation modes: batch, fed-batch and continuous. Cell<br />

growth and glucose consumption were assessed as main culture<br />

parameters.<br />

Material and methods: Cell lines: BA/F3 p210 and BA/F3 p210 eIF5A-2<br />

(BA/F3 = mouse pro B cells, p210 = Bcr-Abl oncoprotein (210kDa), eIF5A-<br />

2 = isoform of the eukaryotic translation initiation factor eIF5A).<br />

In a first step, a working cell bank was established and cell growth was<br />

characterized in T-flasks. Afterwards, different cultivation modes were<br />

tested in a stirred tank bioreactor (Vario1000, Medorex, Germany) as<br />

follows:<br />

batch: Cultivation volume Vstart = 350 mL, duration: 40 h<br />

fed-batch: Cultivation volume V start = 345 mL, duration: 64 h, Feeding took<br />

place every time Glucose concentration fell below 2 mM. Feed medium<br />

consisted on a mixture of batch medium and higher concentrations of<br />

glucose and glutamine.<br />

Continuous: Cultivation volume V start = 115 mL, dilution rate D = 0.049 h -1<br />

duration: 118.5 h.<br />

Thescale-upexperimentwasperformedina5Lstirredbench-top<br />

bioreactor (Biostat B, Sartorius Stedim Biotech GmbH) with pH and DO<br />

control.<br />

Results and conclusions: In batch mode, the maximum viable cell<br />

density during exponential growth was VCD max = 14.7·10 5 cells mL -1 .In<br />

fed-batch mode VCD max = 22.6·10 5 cells mL -1 . This higher cell density is an<br />

advantage over the batch culture mode. It was not possible to obtain<br />

higher cell densities in this mode, since the feed medium consisted on a<br />

formulation for batch culture with further addition of glucose and<br />

glutamine. In continuous mode the highest possible cell density was<br />

maintained in the bioreactor, in order to produce continuously cells for<br />

further treatment. A maximum cell yield of 8.3·10 6 cells h -1 could be<br />

harvested from the bioreactor. After scale-up, this yield might be<br />

increased, so that the needed cell number could be harvested in only few<br />

days. A disadvantage of the continuous process with cell harvest was<br />

observed for the storage process, since cell lysis took place after storage at<br />

4 °C.<br />

A first approach for scale-up was performed in the 5 L bioreactor (Figure 1),<br />

where the maximum cell density during exponential phase allowed for the<br />

needed cell number. Regarding the required reproducibility for cultivation,<br />

the 5 L batch mode was preferred over T-flasks due to the possibility for<br />

control of process variables like pH and pO 2.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P48) Schematic diagram of the final batch process in a 5 L bioreactor that yields a total cell number close to 1·10 10 .<br />

Figure 2(abstract P48) V start = 5090 mL, max. viable cell density in exponential growth after 39.5 hours VCD max = 18.1·10 5 cells mL - .<br />

Page 74 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Compared to T-flasks, glucose uptake during bioreactor cultivation was<br />

much higher, which led to lower final-cell-density yields. fed-batch and<br />

continuous modes were firstly favored due the theoretical final cell<br />

numbers reached during culture. However, the difference in growth,<br />

limitation of bioreactor volume and the need of a special medium<br />

formulation for higher cell densities during fed-batch, limited the final<br />

yield. Continuous mode with temperature reduction of harvested cells<br />

allowed for constant cell production in exponential phase. On the other<br />

hand, storage of intact cells was limited probably due to protease action.<br />

The 150 mL batch cultivation was scaled up to 5 L in a stirred bench-top<br />

bioreactor (Biostat B, Sartorius Stedim Biotech GmbH).<br />

Reference<br />

1. Balabanov S, et al: Hypusination of eukaryotic initiation factor 5A (eIF5A):<br />

a novel therapeutic target in BCR-ABL-positive leukemias identified by a<br />

proteomics approach. Blood 2007, 109(4).<br />

P49<br />

Physical methods for synchronization of a human production cell line<br />

Oscar Platas Barradas 1 , Uwe Jandt 1 , Ralf Hass 2 , Cornelia Kasper 3 ,<br />

Volker Sandig 4 , Ralf Pörtner 1 , An-Ping Zeng 1*<br />

1<br />

Institute of Bioprocess and Biosystems Engineering, Hamburg University of<br />

Technology, Hamburg, 21073, Germany; 2 Gynecology and Obstetrics. Medical<br />

University Hannover, Hannover, 30625, Germany; 3 Institute for Technical<br />

Chemistry, Leibniz University Hannover, Hannover, 30167, Germany;<br />

4<br />

ProBioGen AG, Berlin, 13086, Germany<br />

E-mail: aze@tuhh.de<br />

BMC Proceedings 2011, 5(Suppl 8):P49<br />

Background: The study of central metabolism and the interaction of its<br />

dynamics during growth, product formation and cell division are key tasks<br />

to decode the complex metabolic network of mammalian cells. For this<br />

purpose, not only the quantitative determination of key cellular molecules<br />

is necessary, but also the variation of their expression rates in time, e.g. cell<br />

cycle dependent gene expression. Thus, synchronization of cultured cells is<br />

a requisite for almost any attempt to elucidate these time dependent<br />

cellular processes. Synchronous cell growth can help to gain deeper<br />

insight into dynamics of cellular metabolism.<br />

In our work, physical methods for synchronization of the human production<br />

cell line AGE1.HN (ProBioGen AG) are experimentally tested. Cell-size<br />

distribution, DNA-content and the number of synchronous divisions are<br />

used for comparison of the methods.<br />

According to our results, the enrichment of an AGE1.HN cell population<br />

within a cell cycle phase is possible. Currently, the increase of cell yield and<br />

the improvement of conditions for cell-growth resumption after<br />

synchronization are being studied.<br />

Synchronization methods: Synchronization of cells can be defined as the<br />

enrichment of cells within a certain phase of the cell cycle. Many authors<br />

have pointed to the importance of further synchronous growth or even a<br />

narrow cell size distribution. Thus, a synchronized culture is one in which<br />

cells of similar age progress as a cohort through the division cycle [1].<br />

Physical and chemical methods for cell synchronization are well described<br />

in literature. The choice depends not only on the cell type (suspension,<br />

adherent, growth medium, etc.), but also on the desired yield of cells and<br />

the degree of synchrony. For the analysis of cell-cycle dependent<br />

metabolic processes, the method of choice should not alter the rate of<br />

cellular reactions as they occur normally. The following physical methods<br />

are part of this study: (1) Temperature reduction: cell growth can be<br />

slowed down by means of a reduction of temperature during culture [2],<br />

which allows for enrichment of cell populations within the G 1 and early<br />

Table 1(abstract P49) Flow cytometry analysis of one elutriated fraction (ELU8)<br />

S-phase. Since yields obtained by using this method are low, temperature<br />

cycles can be used for yield improvement. (2) Gradient centrifugation:<br />

cells can be separated according to their density in a gradient by<br />

centrifugation. A sucrose gradient can be used for this purpose, with<br />

the advantage of being a simple and less expensive procedure.<br />

(3) Counterflow centrifugal elutriation: cells are separated in a<br />

centrifugal field; at the same time, a fluid in counterflow is used to<br />

separate the cells according to their mass within the centrifugal field.<br />

Depending on the mass of the cell, cell populations can be eluted out of<br />

the system by increasing the flow rate of the fluid.<br />

Materials and methods: (1) Temperature reduction: shake flasks were<br />

inoculated with 1·10 6 cells mL -1 and cultivated at 37 °C and 5% CO 2. After<br />

24 h the flasks were set at a reduced temperature. Sampling was<br />

performed at least every 24 h. Samples were treated for flow-cytometry<br />

analysis. For yield improvement a repeated-batch strategy with<br />

temperature cycles (37 °C ® 30 °C, 28 °C) was performed in a controlled<br />

bioreactor (1 L, pH = 7.15, DO = 25% air sat.).<br />

(2) Gradient centrifugation: discontinuous sucrose gradients were<br />

prepared in 50 mL centrifuge tubes. A first gradient considered 5 to 60%<br />

(w/w) of sucrose in culture medium. In order to avoid undesired mixing of<br />

the gradient layers during layer addition, the tubes were set every time at<br />

-80°C after each new addition. Phenol red was used for visual identification<br />

of the layers. A second gradient was produced (20 – 50% w/w sucrose),<br />

which approaches to the density range of AGE1.hn cells. 1.5·10 8 cells were<br />

centrifuged and added to the top of the gradient. Centrifugation was<br />

performed at 230 g for 10 min. After centrifugation samples from the<br />

gradient were washed in phosphate saline solution (PBS) and analyzed for<br />

cell-size distribution (Z2 Particle Counter, Beckman Coulter, Germany).<br />

(3) Counterflow centrifugal elutriation: 7·10 8 cells were centrifuged and<br />

resuspended in 5 mL PBS. This volume was inserted under sterile<br />

conditions into an elutriator (Beckman Coulter) and pumped at a minimal<br />

velocity into the separation chamber. Pumping rate of sterile PBS was<br />

increased stepwise in order to elute fractions of cells. Nine fractions were<br />

collected. The size distribution of the fractions and their DNA-content<br />

(flow cytometry) were analyzed. All fractions were resuspended in fresh<br />

medium and cultivated in an incubator.<br />

Results and conclusions: Pre-experiments for temperature reduction<br />

had shown previously an increase of up to 80% of S-Phase DNAcontent<br />

during shake-flask culture at 30 °C. Further temperature<br />

reduction (28 °C) was needed during repeated batch cultivation with<br />

temperature cycles for cell growth arrest. A viability decrease after<br />

temperature resumption (37°C) was observed after 50 h at 28 °C and<br />

might be attributed to apoptosis. Duration of the temperature cycles is<br />

being currently studied.<br />

High viability was observed in all samples after gradient centrifugation.<br />

Cell size distribution was reduced for one sample to almost 80% of cells<br />

between 12.5 and 15 µm. Gradient can be reduced in order to sharpen<br />

the separation of the cells. Further culture of a cell population and cell<br />

cycle analysis are needed to show the feasibility of this method.<br />

By using the Counterflow Centrifugal Elutriation, cells enriched in<br />

different phases of the cell cycle were obtained. The cell-cycle analysis and<br />

the growth curve of one of the fractions (ELU 8) are presented in Table 1<br />

and Figure 1.<br />

Funding by the BMBF, Grand Nr. 0315275A is gratefully acknowledged.<br />

References<br />

1. Cooper S: Reappraisal of G1-phase arrest and synchronization by<br />

lovastatin. Cell Biology International 2002, 26(8):715-727.<br />

2. Enninga IC: Use of low temperature for growth arrest and<br />

synchronization of human diploid Fibroblasts. Mutation Research 1984,<br />

130:343-352.<br />

Cell cycle phase Non-Sync. Elu8<br />

sub G1 1,5 0,6<br />

G0/G1 61,6 5,9<br />

S 17,8 20,8<br />

G2/M 14,2 52,6<br />

An increase of more than 35 % of cells in the G2/M-Phase was observed for this fraction.<br />

Page 75 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P49) 6-day culture of ELU8. Further culture was possible without noticeable perturbation of cell metabolism.<br />

P50<br />

Efficient production of recombinant IgG by the GLUT5 co-expression<br />

system<br />

Yuichi Inoue 1* , Aiko Inoue 2 , Hiroharu Kawahara 1<br />

1 The Cell Engineering Center, Kitakyushu National College of Technology,<br />

Kitakyushu, 802-0985, Japan; 2 KYURIN CORPORATION, Kitakyushu, 806-0046,<br />

Japan<br />

E-mail: inoue@kct.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):P50<br />

A fructose containing cell culture medium has the advantage of low<br />

lactate production and a small pH change, leading to cell and product<br />

stability. But, not all cell lines grow well in the medium, and the fructose<br />

transporter, GLUT5, is related to it [1]. Thus, we developed an efficient<br />

production system of recombinant proteins by metabolic control and coexpression<br />

with GLUT5 in a fructose-based medium [2]. In this report, the<br />

availability of the GLUT5 co-expression system was indicated in CHO-K1<br />

and the human cell line, SC-01MFP [3].<br />

As a model, an IgG and GLUT5 co-expression vector was constructed and<br />

transfected into cells. When the transfected CHO-K1 and SC-01MFP cells<br />

were cultured in the fructose-based medium, both IgG productions were<br />

increased up to about two-fold of that cultured in the glucose-based<br />

medium (Table 1). Our study may be useful for efficient production of<br />

recombinant proteins using the fructose-based cell culture. In particular,<br />

the production in SC-01MFP cells is valuable for functional analysis of<br />

recombinant proteins with a human glycosylation profile.<br />

References<br />

1. Inoue Y, Kawahara H, Shirahata S, Sugimoto Y: Retinoic acid improves a<br />

hybridoma culture in a fructose-based medium by up-regulation of<br />

Table 1(abstract P50) Proliferation and IgG production in the fructose-based medium<br />

fructose incorporation via retinoid nuclear receptors. Biosci Biotechnol<br />

Biochem 2006, 70:2248-2253.<br />

2. Inoue Y, Tsukamoto Y, Yamanaka M, Nakamura S, Inoue A, Nishino N,<br />

Kawahara H: Efficient production of recombinant IgG by metabolic<br />

control and co-expression with GLUT5 in a fructose-based medium.<br />

Cytotechnology 2010, 62:301-306.<br />

3. Kawahara H: Human cell stains for protein production, provided by<br />

selecting strains with high intracellular protein and mutating with<br />

carcinogens. UK Patent 2008, GB2426523.<br />

P51<br />

Innovative animal component-free surface for the cultivation of human<br />

embryonic stem cells<br />

Thomas Stelzer 1* , Tina Marwood 1 , Cindy Neeley 2<br />

1 Thermo Fisher Scientific Labware and Specialty Plastics, Roskilde, DK-4000,<br />

Denmark; 2 Thermo Fisher Scientific Labware and Specialty Plastics, Rochester,<br />

NY 14625, USA<br />

E-mail: Thomas.stelzer@thermofisher.com<br />

BMC Proceedings 2011, 5(Suppl 8):P51<br />

Background: The promise of pluripotent stem cells lies in their ability to<br />

form any cell or tissue in the body. However, this promise requires a<br />

stable and reproducible method to grow the cells. Current methods rely<br />

on feeder cells or extracellular matrix proteins to cover the cultureware<br />

growth surface, and either manual selection or enzymatic dissociation in<br />

cell passaging and harvesting. This study describes a novel and simple<br />

method to grow pluripotent stem cells without the use of feeder cells or<br />

extracellular matrix proteins.<br />

Materials and methods: Human ESC cultivation<br />

Cell line Relative cell proliferation Relative IgG production<br />

CHO-GLUT5/IgG 1.02 ± 0.19 1.77 ± 0.59<br />

SC-01-GLUT5/IgG 0.86 ± 0.01 1.84 ± 0.04<br />

Page 76 of 181<br />

Cells (1 x 10 5 cells/ml) were cultured in the glucose- and fructose-based media. After 3 days, cell proliferation and recombinant IgG production were compared<br />

between two media. Each value in the glucose-based culture is estimated as 1.00. Data represent relative values of means ± SD (n = 3).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P51) A. Expression of pluripotency markers in human ESC as determined by qRT-PCR after 1 passage (p1), 4 passages (p4), and 8<br />

passages (p8) on the Nunclon Vita surface. B. Expression of pluripotency markers in human ESC as determined by flow cytometry after 4 passages (p4)<br />

and 11 passages (p11) on the Nunclon Vita surface. C. Expression of pluripotency markers in human ESC as determined by immunofluorescence staining<br />

after 11 passages on the Nunclon Vita surface.<br />

Cells: Passage-49 human ESC (H1 line from WiCELL, USA) were maintained<br />

in mouse embryonic fibroblast (MEF)-conditioned medium on Nunclon<br />

Delta surface (Thermo Fisher Scientific, USA) coated with a 1:30 dilution<br />

of growth-factor reduced Matrigel (Becton Dickinson, USA). Cells were<br />

dissociated from the surface for passage by treatment with 1 mg/ ml<br />

collagenase, and then seeded onto Nunclon Vita surface with or without<br />

Rho-kinase inhibitor in the medium, as described below.<br />

Cultivation without Rho-kinase inhibition: H1 ESC was grown for 4 passages<br />

in MEF-conditioned medium on Nunclon Vita surface. Cells were dissociated<br />

from the surface for passage by treatment with 1 mg/ml collagenase. Cells<br />

plated on the Nunclon Vita surface took 7 days of culturing before they<br />

were ready for passage.<br />

Cultivation with Rho-kinase inhibition: H1 ESC were grown in MEFconditioned<br />

medium supplemented with Rho-kinase inhibitor, Y-27632 (10<br />

µM unless otherwise indicated; Sigma-Aldrich, USA). Cells were dissociated<br />

from the surface for passage by treatment with 1 mg/ml collagenase. Cells<br />

plated in medium with 10 µm Y-27632 on the Nunclon Vita surface were<br />

ready for passage 4 days after plating. Cells were grown for the number of<br />

passages as indicated.<br />

Human ESC characterization: Colony presence and morphology were<br />

determined using phase-contrast microscopy, and by the naked eye after<br />

staining colonies with 0.5% crystal violet.<br />

Pluripotency was determined by the presence of pluripotency markers<br />

through the use of qRT-PCR for gene expression, flow cytometry for cell<br />

surface marker expression, and immunofluorescence for cell-surface and<br />

nuclear proteins.<br />

Karyotypic stability was determined by cytogenetic analysis of 20 Gbanded<br />

metaphase cells, and by fluorescent in situ hybridization (FISH) on<br />

200 interphase nuclei using probes for the ETV6 BAP (TEL) gene located on<br />

chromosome 12 and for chromosome 17 centromere.<br />

Ability to form embryoid bodies was determined by growing ESC in a lowbinding<br />

plate for 10 days in DMEM/F12 containing 10% FBS.<br />

Results: Human ESCs can be passaged a few times on the Nunclon Vita<br />

surface in MEF-conditioned media without Rho-kinase inhibition before<br />

the growth rate spontaneously declines. Such decline in growth rate of<br />

human ESC on the Nunclon Vita surface is prevented by supplementing<br />

the conditioned medium with Rho-kinase inhibitor Y-27632.<br />

Human ESCs grown on the Nunclon Vita surface in the presence of<br />

Y-27632 have normal karyotype, express pluripotency markers (Fig. 1),<br />

and can be differentiated into embryoid bodies. Non-enzymatic<br />

passaging of human ESCs on Nunclon Vita surface can be accomplished<br />

by removing Y-27632 from the culture media 15 to 30 minutes before<br />

sub-culturing.<br />

Conclusions: The Nunclon Vita surface support feeder cell- and<br />

extracellular matrix-free attachment, colony formation and growth of<br />

human ESC:<br />

For a few passages in media conditioned by mouse embryonic<br />

fibroblasts<br />

For over 10 passages in media conditioned by mouse embryonic<br />

fibroblasts and supplemented with Rho-kinase inhibitor Y-27632. The<br />

human ESCs expanded 11 passages on the Nunclon Vita surface<br />

maintained normal karyotype, pluripotency, and their ability to<br />

differentiate into germ layer cells.<br />

P52<br />

Avian cell line - Technology for large scale vaccine production<br />

Barbara Kraus, Simone von Fircks, Simone Feigl, Sabrina M Koch,<br />

Daniel Fleischanderl, Katherine Terler, Mitra Dersch-Pourmojib,<br />

Christian Konetschny, Leopold Grillberger, Manfred Reiter *<br />

Baxter Innovations GmbH, Uferstrasse 15, 2304 Orth/Donau, Austria<br />

E-mail: manfred_reiter@baxter.com<br />

BMC Proceedings 2011, 5(Suppl 8):P52<br />

Page 77 of 181<br />

Introduction: The establishment of an immortalized continuous cell line<br />

derived from quail cells was undertaken by Baxter in order to develop a<br />

new cell line platform for vaccine production that is free of genetically<br />

modifying sequences.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Important vaccines and viral vectors are still produced in embryonated<br />

chicken eggs or primary chicken embryo fibroblasts. However, the<br />

substitution of primary cells by a continuous cell line has several<br />

advantages. Although primary avian tissue for virus replication is<br />

provided by specific pathogen-free (SPF) production plants, sterility<br />

during vaccine production on embryonated eggs is difficult to guarantee,<br />

and the constant risk of contamination necessitates the addition of<br />

antibiotics. In addition, the supply of embryonated SPF eggs could be a<br />

limiting factor in vaccine production if increased amounts are demanded<br />

by the vaccine manufacturers, e. g. in case of a pandemic outbreak.<br />

Thesameistrueforapproacheswhere primary fibroblast monolayer<br />

cultures are used. Thus, avian cell lines have become a modern option for<br />

vaccine manufacturing and will definitely replace egg and primary<br />

fibroblast technology.<br />

Cell Line Development and Characterization: Here we describe the<br />

development of a continuous avian cell line from quail embryos into<br />

serum-free suspension culture and its manufacturing potential for several<br />

different vaccines.<br />

Briefly, embryos of Colinus virginianus virginianus were disintegrated and<br />

trypsinized to obtain a primary culture that grew adherently in serumcontaining<br />

medium. After UV treatment with a specific dose [1], immortalized<br />

cells were expanded and subsequently adapted to serum-free conditions<br />

(QOR2-sf) and growth in suspension using a chemically defined medium.<br />

Subcloning resulted in the isolation of clone QOR2/2E11, which was selected<br />

as the lead clone for development of vaccine production processes. Then,<br />

quality-controlled cell banks were established (Figure 1). The cell clone<br />

QOR2/2E11 is grown in a chemically defined, animal component-free<br />

medium (Baxter proprietary formulation).<br />

Quality control (QC) testing was performed at different stages during cell<br />

line development. Extended characterization according to relevant<br />

guidelines (Table 1) showed that the cells are free of adventitious agents<br />

and F-Pert negative. Tumorigenicity testing performed in BALB/c nude<br />

mice indicated no tumorigenic effect. Accordingly, the cells fulfill all the<br />

critical regulatory requirements.<br />

Figure 1(abstract P52) Schematic overview of the cell line history.<br />

Page 78 of 181<br />

Thus, the developed subclone QOR2/2E11 was considered to be qualified<br />

as source material for Good Manufacturing Practice (GMP) cell bank<br />

production. A pre master cell bank and cell bank derivates were produced<br />

in compliance with the current GMP regulations and are currently<br />

available.<br />

Scalable live virus production in QOR avian cells: For process<br />

development purposes, modified vaccinia Ankara (MVA) virus was used as<br />

a model virus. MVA virus is a highly attenuated strain of vaccinia virus<br />

belonging to the Poxviridae family that was produced by over 500<br />

passages in chicken embryo fibroblasts. MVA has lost about 10% of the<br />

vaccinia dsDNA genome and consequently cannot replicate in primate<br />

and human cells. Its complex genome of circa 200 kb allows the insertion<br />

of large exogenous DNA inserts. Therefore, MVA serves as a versatile live<br />

vector for the development of human vaccines against diverse disease<br />

targets, such as malaria and cancer, for which conventional approaches<br />

have so far failed [2].<br />

Using the QOR cell line, high product titers can be achieved with a broad<br />

rangeofvirusessuchaswild-typeMVA, recombinant MVA (rMVA) strains,<br />

influenza and flaviviruses. Figure 2 shows the growth of virus in 200 ml<br />

spinner flask cultures using different rMVA constructs. The experiments<br />

were performed with a starting cell density of about 2 x 10 6 cells per ml<br />

and a multiplicity of infection between 1.0 and 0.1. Over the course of four<br />

days post infection (dpi), virus titers of ≥ 1x10 9 TCID 50 per ml were<br />

obtained with every rMVA construct tested. Constant cell performance up<br />

to 10L bioreactors (laboratory scale), where cell densities ≥ 2x10 6 cells per<br />

ml were achieved, has been confirmed and scalability to 100 - 1000L<br />

bioreactors is currently under evaluation.<br />

Furthermore, this avian cell line can serve as a control cell line with<br />

different model viruses in QC tests for adventitious agents.<br />

Conclusion: A scalable technology for the production of live and<br />

attenuated vaccines based on the qualified avian cell line QOR2/2E11 has<br />

been established. The markedly advantages are:<br />

Stable, continuous avian cell line established without the introduction of<br />

foreign genes


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P52) List of guidelines according to which the avian cell line was tested<br />

Organization Number Title<br />

European Directorate for the quality of Medicines &<br />

HealthCare<br />

The International Conference on Harmonisation of<br />

Technical Requirements for Registration of<br />

Pharmaceuticals for Human Use (ICH)<br />

Post production cells are not tumorigenic in animal model<br />

Suspension growth in low-cost chemically defined medium<br />

TCID50 titers ≥ 1x10 9 for several rMVA constructs tested<br />

References<br />

1. Reiter M, et al: Method for producing continuous cell lines. US2009/<br />

0215123 A1 (Patent Application Publication, United States, 2009).<br />

2. Clinical Trials Feeds. [http://clinicaltrialsfeeds.org].<br />

P53<br />

Towards human central nervous system in vitro models for preclinical<br />

research: strategies for 3D neural cell culture<br />

Daniel Simão 1,2 , Inês Costa 1,2 , Margarida Serra 1,2 , Johannes Schwarz 3 ,<br />

Catarina Brito 1,2* , Paula M Alves 1,2<br />

1 Instituto de Tecnologia Química e Biológica –Universidade Nova de Lisboa,<br />

2780-157 Oeiras, Portugal; 2 Instituto de Biologia Experimental e Tecnológica,<br />

European<br />

Pharmacopoeia.<br />

(EP) 2.6.16.<br />

Tests for extraneous agents in viral vaccines for human use<br />

EP 5.2.3. Cell substrates for the production of vaccines for human use<br />

Q5A (R1)-1999 Viral safety evaluation of biotechnology products derived from<br />

cell lines of human of animal origin<br />

Q5D-1997 Derivation and characterization of cell substrates used for<br />

production of biotechnological/biological products<br />

Q7A-2000 Good manufacturing practice guide for active pharmaceutical<br />

ingredients<br />

Center for Biologics Evaluation & Research (CBER) —— Points to Consider in the Characterization of Cell Lines Used to<br />

Produce Biologicals (1993)<br />

—— Guidance for Industry, Characterization and Qualification of Cell<br />

Substrates and Other Biological Materials Used in the Production<br />

of Viral Vaccines for Infectious Disease Indication (February 2010)<br />

Code of Federal Regulations (CFR) 9CFR113.47 Detection of extraneous agents viruses by fluorescent antibody<br />

technique<br />

21CFR610.18 General biological products standards<br />

Figure 2(abstract P52) Growth Kinetics of rMVA viruses on QOR2/2E11.<br />

Page 79 of 181<br />

2780-901 Oeiras, Portugal; 3 Department of Neurology, University of Leipzig,<br />

04103 Leipzig, Germany<br />

BMC Proceedings 2011, 5(Suppl 8):P53<br />

Background: The development of new drugs for human <strong>Central</strong> Nervous<br />

System (CNS) diseases has traditionally relied on 2D in vitro cell models<br />

and genetically engineered animal models. However, those models often<br />

diverge considerably from that of human phenotype (anatomical,<br />

developmental and biochemical differences) [1] contributing to a high<br />

attrition rate - only 8% of CNS drugs entering clinical trials end up being<br />

approved [2]. Human 3D in vitro models are useful complementary tools<br />

towards more accurate evaluation of drug candidates in pre-clinical stages,<br />

as they present an intermediate degree of complexity in terms of cell-cell<br />

and cell-matrix interactions, between the traditional 2D monolayer culture<br />

conditions and the complex brain and can be a better starting point for<br />

the analysis of the in vivo context. Aiming at developing novel 3D in vitro


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

models of the CNS, this work focus on the implementation of long-term<br />

cultures of human midbrain-derived neural stem cells (hmNSC) for the<br />

scalable supply of neural-subtype cells, with a focus on the dopaminergic<br />

lineage, following a systematic technological approach based on stirred<br />

culture systems.<br />

Materials and methods: Cell culture: hmNSC were isolated as previously<br />

reported [3] and routinely propagated in static conditions, on poly-Lornithine-fibronectin<br />

(PLOF) coated plates, in serum-free propagation<br />

medium, containing basic fibroblast growth factor and epidermal growth<br />

factor [3]. hmNSC were cultured in stirred systems in Cultispher S<br />

microcarriers (Percell Biolytica) without coating and coated with PLOF) or<br />

as neurospheres for 7 to 21 days, with media changes every 3-4 days. All<br />

experiments were performed in 125 mL shake flasks (20 mL working<br />

volume), with orbital shaking at 100 rpm. Cultures were maintained at<br />

37°C, in 3% O 2. Double stain viability test: aggregates were collected from<br />

stirred cultures, incubated with fluorescein diacetate (10 μg/mL) and<br />

propidium iodide (1 μg/mL) and observed on a fluorescence microscope<br />

(Leica DMI6000). Aggregate size was measured in pictures taken from<br />

each culture sample using Image J software (NIH), as previously reported<br />

[4]. Dissociation: For microcarrier cultures, Cultispher S was allowed to<br />

settle,washedwithPBSanddigestedwithTrypsin0.05%-EDTA(Gibco).<br />

Cells were collected by centrifugation and counted by trypan blue<br />

exclusion dye. Free cells were counted using the same aliquot.<br />

Aggregates were dissociated with Accutase (Sigma).<br />

Results: The feasibility of culturing hmNSC as 3D structures in stirred<br />

culture systems was assessed by testing two different approaches:<br />

microcarrier technology versus cell aggregated cultures (neurospheres).<br />

For the first strategy, Cultispher S, a collagen-based macroporous<br />

microcarrier was tested for its ability to support hmNSC attachment and<br />

growth. Microcarriers uncoated and coated with PLOF were tested.<br />

hmNSCs were labelled with PKH26 lipophilic dye (red) for detection<br />

Page 80 of 181<br />

purposes and inoculated at 1.5x10 5 cell/mL, in a carrier concentration of<br />

1g/L, corresponding to approximately 125 cell/microcarrier. Monitoring<br />

along 5 days of culture time revealed that the fraction of viable cells found<br />

on the microcarriers was less than 10% of the inoculum, for both uncoated<br />

and PLOF-coated microcarriers, indicating poor microcarrier colonization.<br />

Fluorescence microscopy analysis revealed that hmNSC aggregated in<br />

suspension rather than colonizing Cultispher S microcarriers (not shown).<br />

For the cell aggregate strategy, two inoculum concentrations were tested -<br />

2and4x10 5 cell/mL and aggregate size and number evaluated along<br />

culture time (Figure 1).<br />

The inoculum concentration of 2x10 5 cell/mL was as efficient as 4x10 5<br />

cell/mL in promoting cell aggregation (Figure 1A) whereas it allowed for<br />

lower mean diameters along culture time (362±32 μm at day 14) as<br />

compared to the higher inoculum concentration for which significantly<br />

higher mean diameter and also a wider range of aggregate sizes were<br />

observed (475±103 μm at day 14) (Figure 1B). Moreover, the lower<br />

inoculum concentration avoided the formation of necrotic centres, which<br />

were detected in cultures with an inoculum concentration of 4x10 5 cell/<br />

mL (Figure 1C).Taken together the data presented indicates that 2x10 5<br />

cell/mL is the most favourable inoculum concentration for culture of<br />

hmNSC as aggregates in stirred culture systems.<br />

Conclusions: In this study the feasibility of culturing hmNSC as 3D<br />

structures in stirred culture systems was evaluated. Cell aggregates<br />

(neurosphere) culture, using an inoculum concentration of 2x10 5 cell/mL<br />

was selected as the best strategy, due to the higher cell viabilities and<br />

tightly control of aggregate diameter attained. The implemented 3D<br />

culture system will be applied in the optimization of differentiation of<br />

hmNSC into dopaminergic neurons, astrocytes and oligodendrocytes.<br />

Acknowledgments: The authors acknowledge the FP7 EU project<br />

BrainCAV (HEALTH-HS_2008_222992) and the FCT project PTDC/EBB-BIO/<br />

112786/2009 for financial support.<br />

Figure 1(abstract P53) Effect of inoculum concentration on aggregate concentration (A), aggregate size (B) and viability (C), evaluated using a double<br />

stain viability test: fluorescein diacetate (live, green); propidium iodide (dead, red). Cells were cultured in shake flasks with inoculum concentrations of 2<br />

and 4x10 5 cell/mL. Error bars denote standard deviation of average of 3 independent experiments. *** indicates significant difference (P


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

References<br />

1. Gagliardi C, Bunnell BA: Large animal models of neurological disorders<br />

for gene therapy. ILAR J 2009, 50:128-143.<br />

2. Miller G: Is pharma running out of brainy ideas? Science 2010, 329:502-504.<br />

3. Milosevic J, Schwarz SC, Ogunlade V, Meyer AK, Storch A, Schwarz J:<br />

Emerging role of LRRK2 in human neural progenitor cell cycle<br />

progression, survival and differentiation. Mol Neurodegener 2009, 4:25.<br />

4. Serra M, Brito C, Costa E, Sousa MFQ, Alves PM: Integrating human stem<br />

cell expansion and neuronal differentiation in bioreactors. BMC<br />

Biotechnology 2009, 8:92.<br />

P54<br />

Evaluation of a disposable stirred tank bioreactor for cultivation of<br />

mammalian cells<br />

Alexander Hähnel 1 , Benjamin Pütz 3 , Kai Iding 2 , Tabea Niediek 2 ,<br />

Frank Gudermann 3 , Dirk Lütkemeyer 1,2*<br />

1 Institute for Protein Characterisation, Faculty of Engineering and Mathematics,<br />

University of Applied Sciences, Bielefeld, Germany; 2 BIBITEC GmbH, Bielefeld,<br />

Germany; 3 Institute of Technical Analytics, Faculty of Engineering and<br />

Mathematics, University of Applied Sciences, Bielefeld, Germany<br />

E-mail: dirk.luetkemeyer@fh-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P54<br />

Introduction: Disposable bioreactors are increasingly gaining acceptance<br />

for cell culture applications due to a number of advantages including ease<br />

of use and reduced labour costs, less requirements for utilities such as steam<br />

and purified water. In addition, the system requires no cleaning or cleaning<br />

validation and only a reduced clean room footprint. Integrated ready to use<br />

pre-sterilised disposable sensors for pH and dO 2 monitoring are expected to<br />

reduce the risk of contamination compared to conventional electrodes.<br />

Accordingly, a disposable pre-sterilised and therefore ready-to-use 200 L<br />

bioreactor featuring optical sensors for dO 2 and pH has been evaluated for<br />

suspension culture of CHO cells in protein free media.<br />

Materials and methods: Several fed-batch cultivations with a maximum<br />

cultivation time of 9 days were performed as follows.<br />

Cells: Recombinant CHO cell lines producing monoclonal antibodies<br />

Media: MAM-PF2, Bioconcept, Allschwil, Switzerland<br />

Feed and medium from Teutocell, Bielefeld, Germany<br />

Both media are chemically defined and protein free.<br />

Feeds: Different commercial available amino acid concentrates, glucose<br />

and glutamine<br />

Bioreactor: BIOSTAT® CultiBag STR 200 L, Sartorius Stedim biotech,<br />

Göttingen, Germany<br />

Parameter control: Agitation of the culture was performed by a preinstalled<br />

magnetic driven stirrer with two 3-blade impellers at 80 rpm, pHadjustment<br />

via adding CO 2 to the overlay stream and dO 2 was controlled<br />

via aeration of pure oxygen through the ring-sparger.<br />

Results: Maximum cell densities between 5.0 x 10 6 and 1.3 x 10 7 cells/mL<br />

with viabilities between 85% and 100% were achieved. Results of one<br />

typical fed-batch cultivation are shown in figure 1.<br />

The cell densities depended more on the medium performance and feeding<br />

strategy than on the bioreactor-features itself. At a cell density of 1.3 x 10 7<br />

cells/mLthevolumeflowofpureoxygenwasatarateof0.85L/min.<br />

In regard to the system’s maximum flow rate of 20 L/min, the throughput of<br />

oxygen most likely would not become a bottleneck of even higher cell<br />

densities. Detailed comparison of the optical pH and dO 2 probes with their<br />

conventional counterparts revealed a reliable concordant performance.<br />

Conclusion: The disposable BIOSTAT® CultiBag STR 200 L bioreactor is a<br />

reliable cultivation system for high cell density operations above 10 7 cells/<br />

mL with mammalian cells. The usually installed ring-sparger supplies a<br />

sufficient amount of gas into the liquid phase and still offers adequate<br />

reserves. In direct comparison the optical sensors correlated excellently with<br />

the conventional electrodes. External pH-testing and recalibration is<br />

mandatory to compensate for the drift of the pH-sensor. The reactor’s<br />

overall performance is convincing in this field of operation, which is mainly<br />

due to its ease of use combined with the low utilisation of space. The set up<br />

procedure can be done in less than three hours which is remarkable<br />

compared with stainless steel bio-reactors and the advantage in prevention<br />

of cross-contaminants during changeover of campaigns is preeminent.<br />

Acknowlegement: The supply of materials by Dr. Greller and Mrs. Noack<br />

(Sartorius Stedim Biotech GmbH) is greatly acknowledged.<br />

P55<br />

Controlled expansion and differentiation of mesenchymal stem cells in<br />

a microcarrier based stirred bioreactor<br />

Sébastien Sart 1,2* , Abdelmounaim Errachid 1 , Yves-Jacques Schneider 1,3 ,<br />

Spiros N Agathos 1,2<br />

1 Université Catholique de Louvain / Institut des Sciences de La Vie, Belgium;<br />

2 Laboratory of Bioengineering (GEBI), Place Croix du Sud, 2/19, 1348<br />

Louvain-la-Neuve, Belgium; 3 Laboratory of Cellular Biochemistry, Place Croix<br />

du Sud, 4/5 box 3, 1348 Louvain-la-Neuve, Belgium<br />

E-mail: sebastien.sart@gmail.com<br />

BMC Proceedings 2011, 5(Suppl 8):P55<br />

Introduction: Cell based therapy requires great numbers of cells in a<br />

functional state permitting their in vivo implantation for the restoration of<br />

Figure 1(abstract P54) Cell growth and viability during a fed-batch cultivation of CHO cells in the 200 L disposable bioreactor.<br />

Page 81 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

tissue homeostasis. Three main parameters are believed to be essential<br />

for such a purpose: an appropriate cell population, a suitable scaffold and<br />

appropriate physical / biochemical factors enabling proper expansion and<br />

in vitro cell differentiation. In recent years, mesenchymal stem cells<br />

(MSCs) have been attracting a lot of interest in this field, because of their<br />

differentiation potential and their trophic factor secretion abilities. The<br />

aim of this work is to perform a rational analysis of key factors involved<br />

in the efficient proliferation and differentiation of MSCs, in the context of<br />

a stirred microcarrier (MC)-based bioreactor.<br />

Materials and methods: MSCs from external ear (E-MSCs) and bone<br />

marrow stroma (BM-MSCs) were extracted from Wistar rats, selected and<br />

cultivated on plastic dishes as previously described [1]. The differentiation<br />

potential of E-MSCs along the adipogenic, osteogenic and chondrogenic<br />

pathways was established and assessed by staining (respectively Oil red O,<br />

von Kossa, Alcian Blue) as well as by RT-PCR analysis on marker genes of<br />

differentiation (respectively, C/EBPa, osteocalcin, aggrecan) as previously<br />

described [1]. MCs (i.e. Cultispher-S, Cytodex-3, Cytopore-2) were prepared<br />

and cells were seeded as reported in [2]. Cell counting was performed as<br />

follows: (1) after a full digestion of Cultispher-S by trypsin and using trypan<br />

blue exclusion counting method, as in [2]; (2) by crystal violet staining and<br />

nuclei counting for Cytodex-3, as in [2]; or (3) cell counting on Cytopore-2<br />

was performed using MTT according to [3]. The multiplication ratio was<br />

calculated as defined elsewhere [2]. Cell cycle was analyzed by FACS, after<br />

cell staining with propidium iodide, as in [2]. The actin organization was<br />

assed by confocal microscopy after cell staining with phalloidin-rhodamine.<br />

Results:<br />

MSC and microcarrier screening: E-MSCs were compared to the “gold<br />

standard” BM-MSCs on the basis of their proliferative properties. E-MSCs<br />

bear characteristics of progenitor cells: expression of CD73, Sca-1 and<br />

Notch-1, and also in vitro differentiation potential into mesodermal cell<br />

types such as adipocytes, chondrocytes and osteoblasts (not shown).<br />

Thus, these cells are in vitro functionally analogous to BM-MSCs. This cell<br />

population was further selected on the basis of its high intrinsic<br />

proliferation potential in monolayer culture, a clear advantage in the field<br />

of MSC bioprocessing (Table 1).<br />

Next, we analyzed these cells’ behavior on various types of MCs.<br />

Interestingly, both cell types (E- and BM-MSCs) had similar proliferation<br />

profiles under all the conditions tested: Cultispher-S>Cytodex-<br />

3>Cytopore-2 (Table 1). This validated that E-MSCs are a valuable model<br />

for studying MSCs activities on MCs, given their faster growth and easier<br />

handling compared to BM-MSCs (Table 1). In addition, Cultispher-S turned<br />

out to be the most efficient MC for MSC expansion (Table 1).<br />

Maximization of MSC proliferation in MC-based stirred bioreactors:<br />

According to Table 1, a batch culture mode was not sufficient to<br />

promote efficient E-MSC propagation on Cultispher-S. Conversely, cyclic<br />

fed-batch increased E-MSC growth span (Table 1). In addition, the use of<br />

Page 82 of 181<br />

high levels of growth factors (using a pulsed culture composed of 40%<br />

FBS and 1 ng/mL of TGFb1)increasedgrowthspan(Table1).The<br />

beneficial effects of cyclic fed-batch and pulsed culture were linked to a<br />

sustainment of the percentage of cells in S-phase of the cell cycle<br />

compared to batch culture (Table 1). These results underline that the<br />

control of growth factor levels in the medium is the key to maximize<br />

E-MSC growth extent.<br />

Sequential proliferation and differentiation in MC-based stirred<br />

bioreactors, modulating actin organization: We have previously<br />

shown that the addition of differentiation media significantly diminished<br />

E-MSC proliferation. This indicated that the differentiation of E-MSCs on<br />

MCs must be performed sequentially, after an initial proliferation phase.<br />

According to Figure 1.b, after a first step of E-MSC expansion on MCs, it<br />

was shown that the repression of fibrillar actin (adding Y-27632 to the<br />

differentiation medium) maximized adipogenic differentiation on<br />

Cultispher-S, while the promotion of stress fibers (using lysophosphatidic<br />

acid, LPA) diminished it. In the same vein, Y-27632 improved E-MSC<br />

osteogenic differentiation, while LPA lowered the expression of<br />

osteocalcin (Figure 1.c). Cytopore-2, previously shown to promote<br />

disorganized actin form (similar to that of aggregate cultures and E-MSCs<br />

treated with cytochalasin-D), enabled an efficient E-MSC chondrogenic<br />

differentiation, in comparison to Cultispher-S (Figure 1.d). This latter MC<br />

was previously found to enable a proper E-MSCs actin organization<br />

(composed of mixed cortical and fibrillar actin) linked with their efficient<br />

propagation. These results indicate that a tight control of the E-MSC<br />

microenvironment leading to adapted actin shape is the key towards<br />

efficient MSC differentiation on MCs.<br />

Conclusions: According to these data, it emerges that a correct control<br />

of MSC microenvironment in terms of MC composition is necessary to<br />

promote these cells’ efficient proliferation via proper actin organization.<br />

An efficient MC system must also be combined with adapted biochemical<br />

signaling. Indeed, the growth factor content is an essential factor to<br />

monitor towards improved MSC growth yield. As we observed that the<br />

differentiation step could not be combined with expansion, sequential<br />

phases are required for the mass scale production of a given MSC<br />

differentiated phenotype. Similarly to the expansion phase, the microenvironment<br />

to which MSCs are exposed modulates the efficiency of<br />

their differentiation. According to our results, the promotion of an<br />

adequate actin organization is one of the essential parameters enabling,<br />

in association to biochemical signaling from the differentiation medium,<br />

efficient MSC differentiation on MCs.<br />

Taken together, these results open the way toward mass scale production<br />

of MSCs suitable for future in vivo applications.<br />

Acknowledgments: This work was supported by a FSR grant of<br />

Université Catholique de Louvain, and an IN.WALLONIA-BRUSSELS<br />

INTERNATIONAL (IN.WBI) grant.<br />

Table 1(abstract P55) Multiplication ratios of E-MSCs and BM-MSCs on various culture systems. Multiplication ratios of<br />

E-MSCs and percentage of cells in S-phase at day 5 of a 7 day run, under various modes of culture<br />

MSC and MC screening<br />

BM-MSCs Culture system Multiplication ratio<br />

T-Flasks 0.4 ± 0.2<br />

Cultispher-S 0.16 ± 0.1<br />

Cytodex-3 -0.1 ± 0.12<br />

Cytopore-2 -0.5 ± 0.04<br />

E-MSCs T-Flasks 2.4 ± 0.1<br />

Cultispher-S 2 ± 0.3<br />

Cytodex-3 0.7 ± 0.6<br />

Cytopore-2<br />

Maximization of MSC proliferation<br />

-0.6 ± 0.3<br />

E-MSCs on Cultispher-S Mode of culture Multiplication ratio % of cells in S-phase at day 5<br />

Batch 1.5 ± 0.3 1.4 ± 0.3<br />

Cyclic-fed-batch 2.6 ± 0.2 8 ± 1<br />

Pulsed culture 3 ± 0.04 15 ± 1


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P55) RT-PCR analysis of E-MSC differentiation. Spontaneous marker gene expression after MC expansion (control) (a); adipogenic<br />

differentiation (C/EBPa as a marker) after MC expansion, adding an actin modulator in the differentiation medium (Y-27632 or LPA) (b); osteogenic<br />

differentiation (osteocalcin as a marker) after MC expansion, adding an actin modulator in the differentiation medium (Y-27632 or LPA); chondrogenic<br />

differentiation (aggrecan as a maker) after MC expansion, comparing Cultispher-S to Cytopore 2 (d).<br />

References<br />

1. Sart S, Schneider YJ, Agathos SN: Ear mesenchymal stem cells: an efficient<br />

adult mutipotent cell population fit for rapid and scalable expansion.<br />

J Biotechnol 2009, 139(4):291-299.<br />

2. Sart S, Schneider YJ, Agathos SN: Influence of culture parameters on ear<br />

mesenchymal expanded on microcarriers. J Biotechnol 2010,<br />

150(1):149-160.<br />

3. Pabbruwe MB, Stewart K, Chaudhuri JB: A comparison of colorimetric and<br />

DNA quantification assays for the assessment of meniscal<br />

fibrochondrocyte proliferation in microcarrier culture. Biotechnol Lett<br />

2005, 27(19):1451-1455.<br />

P56<br />

3D6 and 4B3: Recombinant expression of two anti-gp41 antibodies as<br />

dimeric and secretory IgA<br />

David Reinhart 1,2* , Robert Weik 2 , Renate Kunert 2<br />

1 Department of Biotechnology, Institute of Applied Microbiology, University<br />

of Natural Resources and Life Sciences, Vienna, Austria; 2 Polymun Scientific<br />

Immunbiologische Forschung GmbH, Vienna, Austria<br />

E-mail: David.Reinhart@boku.ac.at<br />

BMC Proceedings 2011, 5(Suppl 8):P56<br />

Background: Sexually transmitted diseases are predominantly acquired<br />

via mucosal membranes of the rectal or genital tract during sexual<br />

intercourse. This major port of virus entry is naturally defended by the<br />

humoral immune response, with immunoglobulin A (IgA) as the primary<br />

antibody class to elicit mucosal immunity. Dimeric IgA (dIgA) reaches the<br />

luminal side of mucosal tissues by transcytosis through epithelial cells<br />

lining the mucosa. In a first step, dIgAs specifically bind to the basolaterally<br />

expressed polymeric immunoglobulin receptor (pIgR) on epithelial cells.<br />

For release of IgAs on the luminal side the extracellular portion, termed<br />

Page 83 of 181<br />

secretory component (SC), remains attached to the antibody to form<br />

secretory IgA (sIgA) [1,2].<br />

One example of a sexually transmitted disease is the human immunodeficiency<br />

virus (HIV) which annually infects several million individuals on a<br />

global scale and potentially leads to the acquired immunodeficiency<br />

syndrome (AIDS). Although current therapies can reduce disease progression<br />

in infected individuals, no cure is yet available or within reach in near future.<br />

As a consequence increased attention is now being paid to develop drugs<br />

that could prevent virus acquisition.<br />

3D6 and 4B3 are two monoclonal antibodies (mAb) which have originally<br />

been isolated as IgG1 isotype from seroconverted HIV-1 patients and bind<br />

to the principal immunodominant domain of gp41. In the course of this<br />

project both mAbs were isotype switched to IgA1. Recombinant CHO cell<br />

lines were established for the production of 3D6 and 4B3 as dimeric as<br />

well as secretory IgA. While dIgA were expressed by a single cell line, sIgA<br />

are produced by a biochemical association of dIgA with SC. Both dIgA and<br />

sIgA variants were characterized and the contribution of the heavily<br />

glycosylated SC on IgA stability will be investigated.<br />

Antibody expression: MAbs 3D6 and 4B3 (IgG1) were developed at the<br />

Institute of Applied Microbiology [3,4]. Isotype switching was performed<br />

by substitution of the original heavy chain constant region with that for<br />

IgA1 (GenBank Accession Number 184743). For their recombinant<br />

expression as dIgA three plasmids were generated containing the coding<br />

region of either heavy chain, light chain or joining (J) chain. The latter<br />

one should increase dimerization of IgA monomers. Recombinant CHO<br />

clones were selected whereas high producers were isolated by applying<br />

the dihydrofolate reductase (dhfr) system.<br />

Assembly of 3D6 and 4B3 as sIgA is intended by an in vitro biochemical<br />

association of dimeric IgA with human secretory component (hSC). Thus,<br />

CHO cell lines which solely express hSC were established by cotransfection<br />

of two plasmids containing the coding region for hSC and<br />

dhfr, respectiviely.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Cultivation temperature greatly influences productivity: Mammalian<br />

cells are commonly cultivated at 37°C. In order to investigate the effect on<br />

mAb secretion, clones expressing 3D6-dIgA and 4B3-dIgA were propagated<br />

at sub-physiological temperatures. It was observed that temperature<br />

markedly impacts final IgA quantities in culture supernatants. Product titers<br />

of clones expressing 3D6-dIgA were almost 3-fold increased when<br />

incubated at 33 °C as compared to 37 °C. Furthermore, cultivation of 4B3dIgA-expressing<br />

clones at 33 °C, rather than at 37 °C, resulted in end titers<br />

which were approximately 13-fold increased.<br />

Purification of recombinant dIgA: The two established mAb-expressing<br />

cell lines secrete dIgA at different quantities: The best clone producing 3D6dIgA<br />

achieved a mean specific productivity of 59.1±14 pg/cell*day (pcd).<br />

Conversely, the highest 4B3-dIgA secreting clone reaches a mean specific<br />

productivity of 0.6±0.4 pcd. Hence, an increased amount of host cell<br />

proteins is present in cell culture supernatants of 4B3-dIgA as compared to<br />

3D6-dIgA. Therefore, we elaborated a purification protocol which allows the<br />

recovery of both dimeric IgAs at a greatly enhanced purity. The purification<br />

procedure is shown in table 1.<br />

Formation of secretory IgA: Secretory IgA can be assembled in vitro<br />

due to the natural affinity of dimeric IgA for secretory component and<br />

vice versa (5). Initially, supernatants from recombinant cell lines expressing<br />

hSC, 3D6-dIgA or 4B3-dIgA were buffer exchanged against PBS.<br />

Subsequently, sIgA formation was performed by incubation of hSC with<br />

dIgA at 25°C for 3 hours at different molar ratios. However, applying a<br />

molar ratio of 1:1 is commonly described as optimal [5]. The association<br />

of 3D6-dIgA or 4B3-dIgA with hSC was successfully verified by SDS-PAGE<br />

following Western blotting (data not shown). Conjugation of the<br />

immunoblot was performed with a mouse anti-human secretory<br />

component antiserum (Sigma) and detected via an anti-mouse IgG1<br />

antiserum (Sigma).<br />

Conclusions: The anti-HIV-1 mAbs 3D6 and 4B3 were isotype switched<br />

and could successfully be expressed as dIgA in stably transfected CHO<br />

cells. Furthermore, continuous product secretion was obtained for at least<br />

20 passages in spinner flasks.<br />

Cultivation of the recombinant cell lines at different temperatures revealed<br />

that their product expression can markedly be influenced: 3D6-dIgA<br />

expression was nearly triplicated by shifting the temperature from 37 °C to<br />

33 °C. Applying the same conditions, 4B3-dIgA product secretion was nearly<br />

13-fold increased.<br />

A purification protocol was developed which allows the recovery of the<br />

dIgAs 3D6 and 4B3 in highly pure fractions. Furthermore, the elaborated<br />

scheme enables the isolation of dimeric 3D6 from its various high<br />

molecular weight isotypes.<br />

Secretory IgA of both 3D6 and 4B3 can successfully be produced by<br />

mixing dimeric IgA with human secretory component.<br />

Acknowledgement: Funding from the European Community’s Seventh<br />

Framework Programme (FP7/2002-2013) under grant agreement N°<br />

201038, EuroNeut-41.<br />

References<br />

1. Snoeck V: The IgA system: a comparison of structure and function in<br />

different species. Vet Res 2006, 37(3):455-67.<br />

Page 84 of 181<br />

2. Bonner A, Perrier C, Corthésy B, Perkins SJ: Solution structure of human<br />

secretory component and implications for biological function. J Biol<br />

Chem 2007, 282(23):16969-80, Epub 2007 Apr 11.<br />

3. Buchacher A, Predl R, Strutzenberger K, Steinfellner W, Trkola A,<br />

Purtscher M, Gruber G, Tauer C, Steindl F, Jungbauer A, et al:<br />

Generation of human monoclonal antibodies against HIV-1 proteins;<br />

electrofusion and Epstein-Barr virus transformation for peripheral<br />

blood lymphocyte immortalization. AIDS Res Hum Retroviruses 1994,<br />

10(4):359-69.<br />

4. Kunert R, Rüker F, Katinger H: Molecular characterization of five<br />

neutralizing anti-HIV type 1 antibodies: identification of nonconventional<br />

D segments in the human monoclonal antibodies 2G12 and 2F5. AIDS<br />

Res Hum Retroviruses 1998, 14(13):1115-28.<br />

5. Crottet P, Corthésy B: Secretory component delays the conversion of<br />

secretory IgA into antigen-binding competent F(ab’)2: a possible<br />

implication for mucosal defense. J Immunol 1998, 161(10):5445-53.<br />

P57<br />

Creating new opportunities in process control through radio frequency<br />

impedance spectroscopy<br />

Daniel W Logan * , John P Carvell, Matthew P H Lee<br />

Aber Instruments Ltd., Aberystwyth, SY23 3AH, UK<br />

E-mail: daniel@aberinstruments.com<br />

BMC Proceedings 2011, 5(Suppl 8):P57<br />

Introduction: Process control systems are a valuable tool in the research,<br />

development, and production stages of biopharmaceuticals. They allow<br />

cell culture processes to optimise product production through monitoring<br />

and controlling of the inputs to the system (e.g. air, temperature, feeds).<br />

Process control systems usually read an online monitor and adjust the<br />

inputs of the system to keep that monitor at a set point.<br />

The challenge with many process control systems is the inability to<br />

monitor the most informative variables online. Typically, surrogate<br />

variables such as pH and dissolved oxygen are monitored online and<br />

assumed to track changes in cell growth and product production.<br />

Although the simplicity of these types of probes has led to their wide use<br />

in most bioreactors, their use as effective process control drivers is limited<br />

by the assumptions required to link, for example, oxygen uptake rate to<br />

product formation.<br />

The ideal process control variable would be a direct measure of the desired<br />

product which is often difficult to monitor. However, in biopharmaceutical<br />

applications only the cells in the bioreactor are directly responsible for the<br />

production of the desired products. Online monitors which measure the cell<br />

number (or volume) offer a superior method for tracking cell/product<br />

growth and can be used to control and optimise the process control. Radio<br />

frequency impedance spectroscopy is one such method that offers an<br />

opportunity to monitor cell growth directly online and adjust the<br />

fermentation process accordingly.<br />

Background of radio frequency spectroscopy: Radio frequency (RF)<br />

impedance spectroscopy is the most robust method for measuring viable<br />

Table 1(abstract P56) Purification scheme developed for isolation of 3D6 and 4B3 dIgA<br />

STEP EQUIPMENT PROCEDURE RECOVERY<br />

3D6 4B3<br />

dIgA dIgA<br />

Ultra/Diafiltration Kvick Start UDF Cassette, Harvested cell culture supernatant containing dIgA is concentrated and buffer >95 >95<br />

Millipore, 30 kD<br />

exchanged against PBS, pH 7.4<br />

% %<br />

Lectin Affinity Immobilized Jacalin, UDF retentate is immobilized onto a lectin affinity resin. After a washing step, 98.7 106.1<br />

Chromatography Thermo Scientific bound product is eluted by applying 1.5 M D-galactose in PBS, pH 7.4 % %<br />

Ultra/Diafiltration Kvick Start UDF Cassette, The eluate is buffer exchanged against 20 mM Tris, 10 mM NaCl, pH 8.5 >95 >95<br />

Millipore, 30 kD<br />

% %<br />

Anion Exchange DEAE Sepharose FF, GE The retentate is applied onto the anion exchanger. Post washing with 100mM 88.4 86.7<br />

Chromatography Healthcare NaCl product is eluted from the resin using 20 mM Tris, 200 mM NaCl, pH 8.5 % %<br />

Hydrophobic<br />

Interaction<br />

Phenyl-Sepharose 6FF<br />

low sub, GE Healthcare<br />

(NH4) 2SO4 is added to 0.75 M for 4B3-dIgA or 1.25 M for 3D6-dIgA to precipitate<br />

host cell proteins but not mAb itself. After a washing step 3D6-dIgA is eluted<br />

44.1<br />

%<br />

59.1<br />

%<br />

Chromatography<br />

with 0.4 M (NH4)2SO4, which allows to isolate dimeric IgA from other IgA<br />

isoforms. 4B3-dIgA desorbs at 0 M (NH4)2SO4


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

biomass online [1]. RF impedance spectroscopy measures the passive<br />

electrical properties of cells in suspension through the cells’ interaction<br />

with RF excitations; a technique commonly known as dielectric<br />

spectroscopy.<br />

Viable cells are composed of a conducting cytoplasm surrounded by a<br />

non-conducting membrane suspended in a conducting medium. When<br />

an alternating current is applied to the suspension, each cell becomes<br />

polarised and behaves electrically as a tiny spherical capacitor. The<br />

suspension’s reaction to the current is expressed as its permittivity can<br />

be measured by its capacitance and conductivity as a function of<br />

frequency. Viable cells possess intact membranes which prevent the free<br />

flow of ions and allow the cells to polarise. Dead, porous cells and<br />

debris lack an enclosing membrane and are unable to build up charge<br />

separation. Hence, dielectric spectroscopy only measures viable cells<br />

and is immune to both lysed cells and other debris (e.g. carriers) in the<br />

suspension.<br />

At low excitation frequencies the cells fully polarise and the capacitance of<br />

the suspension is maximised. As the excitation frequency increases, the cells<br />

lose their ability to fully polarise and the measured capacitance drops,<br />

eventually the cells have no polarisation at high frequencies. This relaxation<br />

is called the b-dispersion and has been modelled mathematically by the<br />

Cole-Cole function [2]. Analysis of the dispersion provides estimates of the<br />

biomass volume (proportional to cell number density x cell diameter 4 ),<br />

average cell diameter, and the internal conductivity of the cells.<br />

Futura biomass monitor: The Futura biomass monitor is an online, in-situ<br />

instrument that requires minimal setup and no user interaction during<br />

operation. The Futura range consists of biomass monitors tailored to fit<br />

bioreactors from 100mL to over 1000L as well as various disposable systems.<br />

Unlike dissolved oxygen probes which monitor cell growth indirectly<br />

through the oxygen uptake rate of the suspension, Futura measures the<br />

capacitance created directly from the cells. The modularity of the system<br />

allows a Futura to connect to a variety of probe lengths and styles for varied<br />

applications and its hub based system allows multiple instruments to<br />

connect to a single PC and/or PLC.<br />

Comparison of Futura results to offline measurements: The difficulty<br />

with offline measures of biomass growth is the sparse collection rate (often<br />

Page 85 of 181<br />

1 reading/day), the accuracy of the readings, and the difficulty in defining<br />

the difference between viable and non-viable cells. An online biomass<br />

monitor minimises many of these challenges and gives a more complete<br />

view of the suspension progress.<br />

In one example, a cell culture process with CHO cells was monitored online<br />

using a standard remote Futura with Futura SCADA. Additional offline<br />

monitoring including the cell diameter, cell number density, and oxygen<br />

uptake rate also was collected. The online monitoring of biomass shown in<br />

Figure 1 clearly illustrates the growth, stationary, and decline stages of the<br />

cell culture than the offline measurements alone.<br />

Because RF spectroscopy allows differentiation between biomass and cell<br />

size, additional analysis of the cell culture growth curve can be used to<br />

determine optimum parameters. In the figure, the calculated online cell<br />

diameter correlated well with the offline data up to 250 hours but<br />

thereafter there was a sudden dramatic drop in the on-line measurement.<br />

This change is an artefact due to a large shift in the critical frequency but<br />

it provides a key signpost of changes in the cell that are not easily<br />

picked up by the RFI signal alone.<br />

Once a correlation is determined between biomass or cell size and the<br />

production of the desired product, RF spectroscopy can be used to guide<br />

the timing of feeds or adjust the properties of the medium (e.g. oxygen<br />

content) to optimise the cell culture environment.<br />

Conclusions: Monitoring a cell culture process with online RF<br />

spectroscopy creates new opportunities to understand cellular changes<br />

throughout the length of the process. Monitoring the cells directly<br />

reduces the ambiguity in interpreting secondary monitors of both the<br />

cells and the medium as such monitors can lag the production<br />

mechanism or only be indirectly related to the production of the desired<br />

product.<br />

Acknowledgements: The authors gratefully acknowledge Gary Finka for<br />

data used in this paper.<br />

References<br />

1. Logan D, Carvel J: A biomass monitor for disposable bioreactors.<br />

BioProcess International 2011, 9:48-54.<br />

2. Cole K, Cole R: Dispersion and absorption in dielectrics I. Alternating<br />

current characteristics. J of Chem Phys 1941, 9:341-351.<br />

Figure 1(abstract P57) Online and offline measurements of a CHO cell culture process. Top: online capacitance (solid line) and normalised ViCell biomass<br />

volume (crosses). Bottom: estimated average cell diameter from online capacitance (solid line) and ViCell system (crosses).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

P58<br />

Applications of nanoparticels in molecular and cellular biology and<br />

cancer research<br />

Katya Simeonova 1* , Ganka Milanova 2<br />

1<br />

Institute of Mechanics, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;<br />

2<br />

University of Architecture, Civil Engineering and Geodesy, 1000 Sofia,<br />

Bulgaria<br />

E-mail: katyas@bas.bg<br />

BMC Proceedings 2011, 5(Suppl 8):P58<br />

Background: Discovering of carbon nanotubes (CNTs), by S. Iijima in 1991,<br />

[1] was a revolution in nanoscience (nanomaterials) and nanotechnology.<br />

Moreover these nanoscale materials possess perfectly physicomechanicanical,<br />

electronic, optical, properties. They find applications in<br />

technique, engineering, electronics, optoelectronis, space and environments,<br />

[2]. Recently has been established that nanomaterials, play an<br />

important role in molecular and cellular biology and medicine. The aim of<br />

the work presented could be formulated as follows: to discuss some basic<br />

articles, devoted to applications of nanoparticles, nanotechnology based<br />

on gold nanoparticles for cancer research.<br />

Materials and methods: Synthesis methods for nanoparticles (nanoshells,<br />

nanorods, nanocrystals) have been analyzed. Application of gold<br />

nanoparticels for detection and therapy of cancer has been given too, [3].<br />

Nanotechnology has been determined as an interdisciplinary science<br />

combining physics, mechanics, chemistry, materials science, engineering,<br />

biology becames a very good potential in many different fields of<br />

technique, for cancer therapy, in molecular and cellular biology.<br />

Methods for synthesis of nanoparticles: Some methods for synthesis of<br />

nanoparticles: by controlled different reducing agent; a two- phase method<br />

using other reductants; biocompatible bock polymers. Deposition process<br />

(DP), has been applied for synthesis also. It has been established that rods<br />

wires, multi-concentric shells, hollow tubes,capsules,monocrystasetc.<br />

possess exceptional optical and electronic properties. In [4], the Drude<br />

method (model) for description of optical properties is:<br />

2<br />

’ wp<br />

e = 1 −<br />

w + g<br />

2 2<br />

’’ wpg e = 1 −<br />

w w + g<br />

2<br />

2 2 ( )<br />

Figure 1(abstract P58) Dependencies of extinction, versus wavelength for nanoshells with different core radius.<br />

Page 86 of 181<br />

(1)<br />

(2)


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Here: ε′ and ε″ ; ω =2πc/l; l, c,g have been given in [5]. A correlation,<br />

available is:<br />

n<br />

g = gbulk<br />

+<br />

r<br />

F<br />

eff<br />

Here g, gbulk, νF, roff are given in [5]. Both important characteristics for<br />

description of gold nanoparticles, absorption efficiency and scattering<br />

efficiency have been analyzed too. It must be pointed out as well, that<br />

these nanoparticles could employed in the many medical applications.<br />

Results: The paper presented could be considered as a recent review on<br />

application of nanoparticles and nanotechnology in cancer research. In<br />

the work [6], we could find basic research of American scientists in<br />

nanotechnology based for cancer research, (Figure 1).<br />

Conclusions: In conclusions we could say, that paper presented could be a<br />

successful tool for many medical scientists, physicians, molecular biology<br />

scientists, chemists etc. It’s give good knowledge, regarding nanotechnology<br />

based gold nanoparticles for cancer research. Also, some novel<br />

computational models, based on theoretical studies, analyzed here, could be<br />

developed in future inestigations.<br />

Acknowledgments: Authors would like to thank you very much to<br />

Professor Nicole Borth, personally for her cooperation of attending the<br />

ESACT Meeting, in Vienna, Austria from 15-18 May 2011<br />

References<br />

1. Iijima S: Nature. 1991, 395.<br />

2. Simeonova K, Milanova G: A review on the mechanical behavior of<br />

carbon nanotubes (CNTs), as semiconductors. ISCOM2007, Book of<br />

abstracts Peniscola, Spain 2007, 123-136.<br />

3. Weibo C, et al: Applications of gold nanoparticles in cancer nanotechnology.<br />

Review, Nanotechnology, Science and Applications 2008, 27-32.<br />

4. Lihua W, et al: Gold nanoparticle- based optical probes for targetresponsive<br />

DNA structures. Gold Bulletin 2008, 41(1):37-42.<br />

5. Harris N, et al: Tunable integrated absorption by metal nanoparticles: the<br />

case for gold rods and shells. Gold Bulletin 2008, 41(1):5-14.<br />

6. NCI Alliance for Nanotechnology in Cancer. nanoUtah, Piotr Grodzinski,<br />

PhD, Director 2007.<br />

P59<br />

Measurement of sialic acid content on recombinant membrane proteins<br />

Deniz Baycin-Hizal 1* , Sunny Mai 1 , Daniel Wolozny 1 , Ilhan Akan 2 ,<br />

Noboru Tomiya 3 , Karen Palter 2 , Michael Betenbaugh 1<br />

1 Department of Chemical and Biomolecular Engineering, Johns Hopkins<br />

University, Baltimore, MD, 21218, USA; 2 Department of Biology, Temple<br />

University, Philadelphia, PA, 19122, USA; 3 Department of Biology, Johns<br />

Hopkins University, Baltimore, MD, 21218, USA<br />

BMC Proceedings 2011, 5(Suppl 8):P59<br />

(3)<br />

Page 87 of 181<br />

Background: Membrane proteins such as cell adhesion molecules,<br />

receptors, transporters and ion channel proteins all have essential roles in<br />

cell-growth, migration, and flow of information, cell-cell and cell-protein<br />

communication. Membrane proteins are targets of biopharmaceutical<br />

companies because they have diverse effects on the progression of many<br />

diseases [1]. Ion channels are membrane proteins that play critical roles in a<br />

number of cell functions including communication and neuromuscular<br />

activity. Treatment of channelopathy diseasessuchascancer,cardiac<br />

arrhythmia, ataxia, paralysis, epilepsy, memory and learning loss, requires a<br />

broad understanding of ion channel function. Sialic acid is a critical charged<br />

glycan that affects the action potential of potassium channels which leads<br />

to changes in the neuronal system of organisms. In order to understand the<br />

effect of the sialylation on channel function, the presence of the sialic acid<br />

on the protein of interest should be studied. In this study, a novel method<br />

for the quantification of sialic acid is described for a potassium channel<br />

membrane protein.<br />

Materials and methods: Cell lines: HEK293 cells were grown in DMEM<br />

media (Invitrogen, Carlsbad, CA) supplemented with 10% Fetal Bovine<br />

Serum (Invitrogen), 1% nonessential amino acids (Invitrogen) and 1%<br />

L-Glutamine (Invitrogen).<br />

Transfection: Lipofectamine TM 2000 Reagent (Invitrogen) was used to<br />

transfect potassium channel into HEK293 cells. Stable pools were formed<br />

after 15 days of antibiotic selection and 8 different clones were selected<br />

from the pools.<br />

Western blot analysis: Cells were lysed in RIPA buffer containing<br />

complete-mini EDTA free protease inhibitor cocktail (Roche Diagnostics,<br />

Mannheim, Germany). The protein concentrations of each sample were<br />

determined by using a BCA protein assay kit (Pierce, Rockford, IL). Equal<br />

total protein amounts were loaded onto 8% gels and separated by<br />

electrophoresis. The separated proteins were transferred from the gel to<br />

membrane. For detection of the channel protein, membrane was incubated<br />

in 1% milk in PBST solution containing primary antibody with a dilution of<br />

1:1000 overnight at 4°C. A secondary anti-rabbit IgG HRP conjugate<br />

antibody (Amersham, Louisville, CO) was used at a dilution of 1:5000 in 1%<br />

milk/PBST overnight at 4°C.<br />

Immunoprecipitation: The potassium channel protein was purified by<br />

using G-beads coupled antibody.<br />

HPLC analysis: The purified protein was run on the gel and protein band<br />

was cut from the gel. The cut band was acid hydrolyzed to release the sialic<br />

acid. The released sialic acid was derivatized with fluorescent 4,5-<br />

Methylenedioxy-1,2-phenylenediamine dihydrochloride (DMB) reagent and<br />

injected to HPLC [2].<br />

Results: The transient expression of potassium channel was evaluated by<br />

western blot analysis. After 15 days of antibiotic selection, an expression<br />

level of the gene of interest was evaluated by Western blot. The stable pool<br />

expression is an average of the varied expression levels of the protein of<br />

interest in cells. In order to decrease the expression heterogeneity and<br />

Figure 1(abstract P59) Potassium channel sialylation. A) Western blot analysis of channel proteins B) Western blot of the channel protein before and<br />

after sialidase treatment C) Sialic acid presence in HPLC.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

ensure that all the cells contain the same genetic content, eight different<br />

single clonal isolates were selected. The quality and quantity of the protein<br />

expression in these clonal isolates was compared with the protein<br />

expression of stable pool. Figure 1.A shows the western blot of 8 cell clones<br />

and stable pool of potassium channel. Both glycosylated and nonglycosylated<br />

bands of the specific protein can be distinguished in the<br />

Western blot. Although some of the clones and stable pool results showed<br />

non-glycosylated bands and poor expression, clone 1 and 3 showed high<br />

glycosylation quality and quantity. Forthisreason,clone3wasscaledup<br />

and purified using immunoprecipitation. The purified protein was treated<br />

with sialidase and the mobility shift of the protein before sialidase and after<br />

sialidase treatment was compared by running Western blot as shown in<br />

Figure 1.B. Due to the low molecular weight of sialic acid, the mobility shift<br />

detected by Western blot was minimal. In order to quantify the amount of<br />

sialic acid found on the protein, about 6 pmol of purified protein was run on<br />

the SDS-PAGE gel and was cut from the gel. The cut band was acid<br />

hydrolyzed to release the sialic acid and the released sialic acid was<br />

derivatized with DMB reagent and injected into HPLC. The sialic acid<br />

amount that is released from the gel was calculated by using a sialic acid<br />

calibration curve (data not shown). The amount of sialic acid was calculated<br />

to be 19 pmol depending on HPLC and calibration curve results. Since the<br />

potassium channel of interest may have 4 possible sites for the sialylation,<br />

and therefore 24 pmols sialic acid. For this reason, the estimated sialic acid<br />

occupancy efficiency of the channel was found as 79%.<br />

Conclusion: Membrane proteins are to be very difficult to produce and<br />

purify for structural or glycan analysis because of their low expression levels.<br />

In this study, HEK 293 cells were used to express a potassium channel<br />

protein and clonal isolates were picked from stable pools to evaluate the<br />

quality and quantity of the glycosylated protein. A highly expressing clone<br />

with glycosylation was then selected for the purification and sialic acid<br />

analysis. The results showed that the sialic acid occupancy efficiency of the<br />

channel of interest was around 80% when expressed in HEK293 cells.<br />

References<br />

1. O’Connor S, Li E, Majors BS, He L, Placone J, Baycin D, Betenbaugh MJ,<br />

Hristova K: Increased expression of the integral membrane protein ErbB2<br />

in Chinese hamster ovary cells expressing the anti-apoptotic gene BclxL.<br />

Protein Expr Purif 2009, 67:41-47.<br />

2. Hara S, Yamaguchi M, Takemori Y, Nakamura M, Ohkura Y: Highly sensitive<br />

determination of N-acetyl- and N-glycolylneuraminic acids in human<br />

serum and urine and rat serum by reversed-phase liquid chromatography<br />

with fluorescence detection. J Chromatogr 1986, 377:111-119.<br />

P60<br />

Influence of operating parameters of a settling-based perfusion process<br />

on expansion of VERO cells attached on microcarriers<br />

Amal El Wajgali 1 , Frantz Fournier 1 , Eric Olmos 1 , Cécile Gény 2 , Hervé Pinton 2 ,<br />

Annie Marc 1*<br />

1 Laboratoire Réactions et Génie des Procédés, UPR-CNRS 3349, Vandoeuvrelès-Nancy,<br />

France; 2 Sanofi Pasteur, Marcy L’Etoile, France<br />

E-mail: annie.marc@ensic.inpl-nancy.fr<br />

BMC Proceedings 2011, 5(Suppl 8):P60<br />

Background: The growing demand for biologicals produced by animal<br />

cells motivates the development of more efficient and reliable culture<br />

Page 88 of 181<br />

production processes. In the particular case of the industrial production of<br />

viral vaccines by Vero cells adhered on microcarriers, the cell propagation<br />

phases are mainly devoted to reach high cell density in less time while<br />

maintaining a good cell physiological state. Several papers have reported<br />

the optimization of culture conditions for microcarrier cultures [1-4].<br />

Otherwise, perfusion bioreactors, based on continuous medium renewal<br />

and cell retention, can be a good alternative to batch systems. Among the<br />

various technologies for cell retention, gravitational settler is a promising<br />

device for large-scale perfusion culture process. This simple device takes<br />

advantage of the difference between the cell settling rate and the medium<br />

harvesting flow rate. Moreover, in the particularcaseofcellsattachedon<br />

microcarriers, it is more suitable than in the case of single suspended cells.<br />

So, the aim of this work was to evaluate the performances of adherent<br />

Vero cell cultures performed inside a perfused bioreactor using a<br />

gravitational settler as cell retention device. The study focused on the<br />

influence of two operating parameters, such as microcarrier concentration<br />

(MCs) and initial cell density (C0), on the cell growth.<br />

Materials and methods: Vero cells were provided by Sanofi Pasteur and<br />

cultivated in a serum-free medium attached on Cytodex-1 microcarriers<br />

(GE Healthcare). Cultures were performed in a 2 L bioreactor (Pierre<br />

Guérin, France) controlled at pH: 7.2, temperature: 37°C, pO 2: 25% with an<br />

agitation rate of 90 rpm. After 48 h of batch culture, the perfusion<br />

medium flow rate was started at 0.5 vol.d -1 . The harvest flow rate was<br />

made through a settling glass tube. The concentration of adherent cells<br />

was measured by the crystal violet method.<br />

A Design of Experiment (DoE) was set up, with the objective to study the<br />

effect of two operating parameters (MCs and C 0) in order to reach high<br />

maximal cell density and rapid cell growth. The chosen criteria for DoE<br />

response were the maximal cell concentration, either per medium volume<br />

(cells.mL -1 ) or per microcarrier (cells.MCs -1 ), and the population doubling<br />

level (PDL). A D-optimal design was chosen because of the irregularity of the<br />

experimental region. Three levels were defined for each parameter. The<br />

Modde 7 software was used to generate the DoE: 7 experiments plus one<br />

repetition in order to assess the repeatability. Theoretical initial cell densities<br />

(C 0) were corrected by the experimental values. The table 1 gives the<br />

corresponding operating parameters of the various perfused cultures.<br />

Results: The settling tube used as cell retention device was observed to be<br />

not only easy-to-implement but also a reliable system for retention of cells<br />

adhered on microcarriers. Indeed, no microcarrier was found in the harvest<br />

flow rate, whatever the culture operating conditions. The evolution of cell<br />

density was studied for more than 10 days. A final stabilization of the<br />

maximal cell density was observed during several days for all experiments<br />

(Table 1). The repeatability was evaluated with experiments 7 and 8,<br />

performed with the same operating parameters (MCs: 2.5 g.L -1 and C 0:14<br />

200 cells.cm -2 ). Both experiments displayed similar cell kinetics and reached<br />

the same maximal cell density (2.5 x 10 6 cells.mL -1 ).<br />

On the one hand, the experimental kinetics results were compared on the<br />

basis of the maximal cell concentrations. The highest cell density per<br />

medium volume (3.9 x 10 6 C.mL -1 ) was reached for the culture performed<br />

with the highest MCs (5 g.L -1 , exp. 2). But the highest cell density per<br />

microcarrier (300 cells.MCs -1 ) was obtained with the lowest MCs of 1.2 g.L -1<br />

(exp. 3 and 5) whatever the C 0 value (6 000 and 23 500 cells.cm -2 ). It was<br />

also pointed out that, according to the microcarrier concentration used,<br />

the maximal cell density (in cells per medium volume) could depend on<br />

the initial cell-to-bead-ratio. Indeed, the two experiments performed with<br />

Table 1(abstract P60) Operating conditions and experimental results for perfused experiments<br />

Experiments Operating factors Experimental results<br />

MCs (g.L -1 ) C0 (cells.cm -2 ) Cmax (10 6 cells .mL -1 ) Cmax (cells.MC -1 ) PDL<br />

1 2.5 9100 1.9 114 4.2<br />

2 5 23000 3.9 116 2.9<br />

3 1.2 6000 2.5 311 6.3<br />

4 3.8 10100 2.7 106 3.9<br />

5 1.2 28500 2.2 275 3.8<br />

6 3.8 36000 3.2 125 2.4<br />

7 2.5 14200 2.5 149 3.7<br />

8 (7bis) 2.5 14200 2.5 147 4.2


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

2.5 g.L -1 MCs reached 1.9 x 10 6 cells.mL -1 (exp. 1) when C 0 was 9 100 cells.<br />

cm -2 , and 2.5 x 10 6 cells.mL -1 with C 0 of 14 200 cells.cm -2 (exp. 7). The<br />

same conclusion was obtained with 3.8 g.L -1 MCs (exp. 4 and 6). Therefore,<br />

the highest cell concentration was reached when a sufficient number of<br />

cells per microcarrier was respected at the beginning of culture. But, with<br />

1.2 g.L -1 MCs the maximal cell density was similar for the two C 0 used,<br />

suggesting that the maximal cell concentration was less dependent on<br />

initial cell density at very low carrier concentration.<br />

Furthermore, the statistical analysis of the influence of the operating<br />

parameters, on the basis of the three chosen criteria, showed that the best<br />

model was observed with the PDL values. The model predicted correctly<br />

experimental results and was described by a first-order polynomial. The<br />

regression showed satisfactory statistical qualities and neither interaction<br />

nor quadratic effects were found significant. The model indicated that low<br />

values for both operating factors MCs and C 0 favoured a higher PDL.<br />

Conclusion: The easy-to-implement settling tube was a reliable system for<br />

retention of cells adhered on microcarriers. The influence of two operating<br />

parameters (MCs and C 0) on the Vero cell growth was quantified according<br />

to three criteria related to cell growth (cells.mL -1 , cells.MCs -1 and PDL).<br />

While the highest microcarrier concentration led to the highest total<br />

amount of attached cells, the lowest MCs induced the best carrier recovery<br />

by the cells. Moreover both low MCs and C 0 values favored a high PDL.<br />

These results provide a preliminary screening of operating conditions<br />

before undertaking a rational scale-up of the perfusion process.<br />

References<br />

1. Clark JM, Hirtenstein MD: Optimizing culture condition for the production<br />

of animal cells in microcarrier culture. Ann. N. Y. Acad. Sci 1981, 369-33.<br />

2. Mendonça RZ, Prado JCM, Pereira CA: Attachment, spreading and growth<br />

of VERO cells on microcarriers for the optimization of large scale<br />

cultures. Bioprocess Eng 1999, 20:565-571.<br />

3. Sean P, Forestell SP, Kalogerakis N, Behie LA, Gerson DF: Development of<br />

the optimal inoculation conditions for microcarrier cultures. Biotechnol.<br />

Bioeng 2004, 39:305-313.<br />

4. Bock A, Sann H, Schulze-Horsel J, Genzel Y, Reichl U, Möhler L: Growth<br />

behaviour of number distributed adherent MDCK cells for optimization<br />

in microcarrier cultures. Biotechnol. Progr 2009, 25:1717-1731.<br />

P61<br />

3D-Bioreactor culture of human hepatoma cell line HepG2 as a<br />

promising tool for in vitro substance testing<br />

Christiane Goepfert 1 , Wibke Scheurer 1 , Susanne Rohn 1 , Britta Rathjen 1 ,<br />

Stefanie Meyer 1 ,AnjaDittmann 1 , Katharina Wiegandt 2 ,RolfJanßen 2 ,RalfPörtner 1*<br />

1 Institute of Bioprocess and Biosystems Engineering, Hamburg University of<br />

Technology Hamburg, D-21073, Germany; 2 Institute of Advanced Ceramics,<br />

Hamburg University of Technology, Hamburg, D-21073, Germany<br />

E-mail: poertner@tuhh.de<br />

BMC Proceedings 2011, 5(Suppl 8):P61<br />

Page 89 of 181<br />

Introduction: Future developments in pharmaceutical research and<br />

regulatory requirements such as the European REACH program require high<br />

numbers of animal experiments. As a result of ethical concerns, cell culture<br />

tests with human cell lines or primary cells are considered as an alternative.<br />

However, current testing protocols using 2D cell cultures in Petri dishes are<br />

not equivalent to animal trials. 3D tissue cultures may overcome<br />

fundamental obstacles in the development of new therapeutic agents. Many<br />

new candidates of therapeutic agents are intended as agonists or<br />

antagonists of specific receptors on human cells. For these substances,<br />

organ-like test systems based on human cells are mandatory. In some cases,<br />

new pharmaceuticals lead to unexpected adverse reactions even after<br />

successful animal trials. It is assumed that 3D test systems based on human<br />

cells might help to overcome these problems.<br />

Materials and methods: Human hepatoma HepG2 cell line was cultivated<br />

in monolayer culture and on two 3D carrier systems macroporous ceramic<br />

carrier Sponceram (Zellwerk, Germany) and Fibracel composed of polyester<br />

non-woven fiber on a polypropylene scaffold (New Brunswick Scientific,<br />

USA). 3D-carrier cultivation was performed in 24-well-plates, in a multi-well<br />

flow-chamber bioreactor or a fixed bed (Fig. 1) [1]. Cells were seeded at<br />

cell densities of 1*10 5 /ml. Medium was DMEM/Ham´s F-12 mixture<br />

supplemented with 10% FBS. Growth of cultures was determined using<br />

DNA measurement with H33528 after digesting the cells with Papain.An<br />

average DNA content of 14.8 pg DNA per cell was previously obtained<br />

using defined cell concentrations. Functional assays were carried out<br />

according to the method described in [2] using 7-ethoxyresorufin as a<br />

substrate. Induction of EROD activitiy was done using 3-methylcholanthrene<br />

or Ketoconazole. Cellular viability was monitored using<br />

Resazurin and live/dead staining (AO/PI).<br />

Cell growth on carrier systems: On Sponceram, cells spreaded initially<br />

but formed dense clusters after extended cultivation. On FibraCel, cells<br />

formed small aggregates after seeding. Later they grew within the whole<br />

carrier structure. In the flow chamber 3.83 · 10 5 ± 6.04 · 10 4 cells per carrier<br />

were reached within two compared to 1.45 · 10 6 ±3.68·10 6 in 24 well<br />

plates. For the fixed bed an average of approx. 1.35 · 10 6 cells per carrier<br />

were obtained.<br />

Liver specific EROD assay: The cultivation of Hep G2 cells in the two<br />

reactor systems was carried out for 7 days and for 14 days prior to induction<br />

of EROD activity.. Measurement of EROD activity found to be linear for at<br />

least for 1h (sampling every 15 min). Activities were similar to static cultures<br />

in the flow chamber [approx. 1.7 fmol Resorufin/(cell*h)]. Lower activities<br />

were detected in the fixed bed bioreactor after 14d [approx. 0.5 fmol<br />

Resorufin/(cell*h)] compared to 7d [approx. 0.15 fmol Resorufin/(cell*h)],<br />

possibly as a result of the formation of large cell clusters.<br />

Conclusions: In this study it was shown that 3D dynamic culture systems<br />

can be used to carry out functional assays such as the EROD assay with a<br />

human liver cell line. HepG2 cells could be cultivated for a longer time in<br />

dynamic 3D culture and showed a better viability compared to static<br />

monolayer cultivation.<br />

Figure 1(abstract P61) Cultivation systems for 3D-culture of HepG2-cells (A) Multi-well flow-chamber bioreactor (medorex) (B) 10 mL fixed bed reactor<br />

(medorex) [1].


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

References<br />

1. Pörtner R, Platas O, Fassnacht D, Nehring D, Czermak P, Märkl H: Fixed bed<br />

reactors for the cultivation of mammalian cells: design, performance<br />

and scale-up. Open Biotechnol J 2007, 1:41-46.<br />

2. Donato MT, Gómez-Léchon MJ, Castell JV: A microassay for measuring<br />

cytochrome P450IA1 and P450IIB1 activities in intact human and rat<br />

hepatocytes cultured on 96-well plates. Anal Biochem 1993, 213:29-33.<br />

P62<br />

“BioProzessTrainer” as training tool for design of experiments<br />

Ralf Pörtner 1* , Oscar Platas-Barradas 1 , Janosh Gradkowski 1 , Richa Gautam 1 ,<br />

Florian Kuhnen 2 , Volker C Hass 2<br />

1 Institute of Bioprocess and Biosystems Engineering, Hamburg University of<br />

Technology Hamburg, D-21073, Germany; 2 Institute of Environmental and<br />

Bio-Technology, Hochschule Bremen, D-28119, Germany<br />

E-mail: poertner@tuhh.de<br />

BMC Proceedings 2011, 5(Suppl 8):P62<br />

Concept: Design and optimization of cell culture processes requires<br />

intensive studies based on “Design of experiments”-strategies. In<br />

academia teaching of DoE-concepts is often insufficient, as in most cases<br />

only simple culture strategies (batch) can be performed, as time and<br />

money are limited. More complex tasks such as feeding strategies for fed<br />

batch culture can be discussed theoretically only.<br />

To close this gap the virtual “BioProzessTrainer”, a model based simulation<br />

tool, was developed. It supports biotechnological education with respect<br />

to process strategies, bioreactor control, kinetic analysis of experimental<br />

Page 90 of 181<br />

data and modeling. Along with a set of examples for different control and<br />

processstrategies(batch, fed batch, chemostat etc.) learners are prepared<br />

for real experiments [1,2].<br />

The “BioProzessTrainer” (Figure 1) helps to improve the quality of<br />

education by using interactive learning forms and by transmitting<br />

additional knowledge and skills. Costs for practical experiments can be<br />

minimized by reducing plant operation costs. Here a concept for teaching<br />

DoE-concepts for batch- (optimization of e.g. substrate concentrations<br />

and inoculation cell density) and fed-batch-processes (evaluation<br />

and optimization of feeding strategy) using the “BioProzessTrainer” is<br />

shown.<br />

Example 1: DoE for impact of glucose and glutamine concentration during<br />

batch (1,5 L) on cell density and antibody concentration of a mammalian<br />

cell line<br />

Experimental design:<br />

➣ Seed concentration: 4E8 cells/L [±10%]<br />

➣ Glucose conc.: low 15 mmol/L; high 30 mmol/L<br />

➣ Glutamine conc.: low 1 mmol/L; high 4 mmol/L<br />

➣ Culture time: 24h<br />

To induce an experimental error, the seed concentration was varied by +-<br />

10%. Results see Table 1<br />

Analysis via statistical tools:<br />

➣ One-dimensional ANOVA with respect to glucose at high glutamine<br />

concentrations: glucose conc. not significant for cell conc. (p=0.1),<br />

significant for antibody conc. (p=0.044); level of significance 0.05<br />

➣ Two-dimensional ANOVA with repetition: interaction between glucose<br />

and glutamine conc. not significant for cell conc. (p=0.14); significant for<br />

antibody conc. (p=0.046); level of significance 0.05<br />

Figure 1(abstract P62) (A) Teaching material: theoretical back-ground, exercises, sample solution [1] (B) Screen of „BioProzessTrainer“ (C) Example: fedbatch<br />

process with fixed feed rate perfomed with the BioProzessTrainer.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P62) DoE performed with the BioProzessTrainer<br />

cell conc. [ 10 8 cells/L] antibody conc. [mg/L]<br />

seed conc. [10 8 cells/L ] glutamine [mmol/L] glucose [mmol/L]<br />

15 30 15 30<br />

set 4.0 1 7.71 8.01 11.8 12.6<br />

-10% 3.6 1 7.15 7.49 11.1 12.1<br />

+10% 4.4 1 8.23 8.49 12.3 13.0<br />

set 4.0 4 1.17 1.37 22.5 27.7<br />

-10% 3.6 4 1.06 1.24 20.6 25.2<br />

+10% 4.4 4 1.27 1.49 24.4 30.2<br />

Impact of glucose and glutamine concentration on cell density and antibody concentration.<br />

Table 2(abstract P62) Impact of feed rate for glucose and glutamine feed during fed-batch (constant feed rate) on cell<br />

density and antibody concentration<br />

cell conc. [10 9 cells/L] antibody conc. [mg/L]<br />

glutamine feed rate [mL/min] glucose feed rate [mL/min]<br />

0.02 0.08 0.02 0.08<br />

0.02 2.10 2.15 84.2 63.0<br />

0.08 2.95 3.10 67.0 133<br />

Example 2: DoE for impact of feed rate for glucose and glutamine feed<br />

during fed batch (constant feed rate) on cell density and antibody<br />

concentration of a mammalian cell line<br />

Experimental design:<br />

➣ Seed concentration: 8E8 cells/L<br />

➣ Glucose conc. in glucose feed: 180 mmol/L<br />

➣ Glutamine conc. in glutamine feed: 30 mmol/L<br />

➣ Start feed: 24h; start volume 1.5 L; final volume 3 L<br />

➣ Feed rate glucose / glutamine feed: low 0.02 mL/min; high 0.08 mL/min<br />

Results see Table 2<br />

Analysis via statistical tools:<br />

➣ Two-dimensional ANOVA without repetition: glucose feed rate not<br />

significant for cell conc. (p=0.295) and antibody conc. (p=0.699);<br />

glutamine feed rate significant for cell conc. (p=0.035) and not for<br />

antibody conc. (p=0.653); level of significance 0.05<br />

References<br />

1. Hass V, Pörtner R: Praxis der Bioprozesstechnik. Spektrum Akademischer<br />

Verlag978-3-8274-1795-4 2009.<br />

2. Pörtner R, Hass VC: Interactive virtual learning environment for<br />

biotechnology (eLearnBioTec). Chemie-Ingenieur-Technik 2005, 77(8):1256.<br />

P63<br />

ADCC potency assay: increased standardization with modified<br />

lymphocytes<br />

Laurent Bretaudeau * , Véronique Bonnaudet<br />

Clean Cells, Boufféré, 85600, France<br />

E-mail: lbretaudeau@clean-cells.com<br />

BMC Proceedings 2011, 5(Suppl 8):P63<br />

Page 91 of 181<br />

For few years now, the use of monoclonal antibodies represents a<br />

significant progress in different therapeutic applications. In addition to<br />

commercialization of new products, important efforts in research and<br />

development have been made to launch new therapeutic antibodies.<br />

Many antibodies act through a mechanism of Antibody-dependant cell<br />

cytotoxicity (ADCC). National Health Agencies recommend or require the<br />

use of biological activity assays (potency) in order to characterize those<br />

pharmaceutical products. The ADCC assay combines the 3 following<br />

elements:<br />

The antibody of interest that is specific to a given antigen;<br />

The targeted cells that express the antigen of interest at their surface;<br />

Table 1(abstract P63) Advantages and drawbacks of ADCC effectors for the validation of a standardized ADCC assay<br />

ADCC effector types Advantages Drawbacks<br />

Primary NK or PBMC, isolated from Representative from the genetic diversity Not convenient for standardization<br />

donors<br />

Requirement for donor genotyping<br />

Requirement to evaluate several donors for accurate comparison<br />

Exposure of operators to biohazard<br />

Significant ressources required (budget and time) for cell<br />

preparation<br />

Modified NK cell lines Increased suitability for standardization The NK activity may interfere with the measurement of the<br />

ADCC-related lysis<br />

Absence of functionally qualified batches of cells<br />

Absence of biosafety qualified batches of cells<br />

To be handled as a GMO<br />

CD16-transduced lymphocytes Increased suitability for standardization<br />

Qualified batches available in a ready-touse<br />

format<br />

Absence of NK activity-related background<br />

To be handled as a GMO


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P63) Characterization of ADCC function of CD16-transduced lymphocytes. Two models of antigen expressing cells were used to<br />

establish the inter-assay precision of the ADCC measurement in a standard chromium-release assay. Briefly, the cells were 51 Cr-labelled, washed,<br />

incubated with the antibodies and the release of 51 Cr in the supernatant was analyzed after 4h.<br />

The effector that can trigger the lysis the targeted cell when the<br />

antibody is linked to the antigen.<br />

As the usually met ADCC assays use Natural Killer cells, isolated from<br />

healthy donors, as effectors they are hardly reproducible (Table 1). Thus,<br />

those assays can barely be validated when lots of pharmaceutical<br />

products are released. Furthermore, of the rare NK cell lines established<br />

in culture don’t express the CD16 receptor needed for the ADCC function.<br />

In this context, the use of standardized effectors should improve<br />

significantly the ADCC assays. Previous work has highlighted that human<br />

lymphocytes, modified to express the CD16 receptor, have acquired the<br />

ADCC functions [1]. Thanks to our specific know-how, clones of CD16+<br />

lymphocytes have been produced on a large scale (10 9 cellules). The<br />

results obtained have pointed out that cells stored in liquid nitrogen and<br />

used as soon as they were thawed, were usable for ADCC assays on a<br />

reproducible basis (Figure 1). Thanks to that approach, two models of<br />

ADCC measurement are characterized in the present presentation: CD20<br />

and Her2neu.<br />

The produced effector cells constitute a relevant alternative to replace the<br />

use of NK cells when the standardization of ADCC potency assay is<br />

needed.<br />

Reference<br />

1. Clémenceau B, Congy-Jolivet N, Gallot G, Vivien R, Gaschet J, Thibault G,<br />

Vié H: Antibody-dependent cellular cytotoxicity (ADCC) is mediated by<br />

genetically modified antigen-specific human T lymphocytes. Blood 2006,<br />

107(12):4669-77.<br />

P64<br />

Anti-idiotypic antibody Ab2/3H6 mimicking gp41: a potential HIV-1<br />

vaccine?<br />

Renate Kunert * , Alexander Mader<br />

Department of Biotechnology, Institute for Applied Microbiology, BOKU –<br />

University of Natural Resources and Life Sciences, A-1190 Vienna, Austria<br />

E-mail: renate.kunert@boku.ac.at<br />

BMC Proceedings 2011, 5(Suppl 8):P64<br />

Background and aims: Anti-idiotypic antibodies (Abs) represent an<br />

alternative vaccination approach in human therapy. This approach is based<br />

on the idiotype (Id) network theory postulated by Jerne describing the Ab<br />

(Ab1) – anti-idiotypic Ab (Ab2) – anti-anti-idiotypic Ab (Ab3) cascade<br />

stimulation. Specific anti-Id Abs serve as an “internal image” of the target<br />

antigen and can be used to induce Abs able to bind to the cognate<br />

antigen [1]. The anti-Id Ab Ab2/3H6 [2], developed at our Institute was<br />

generated in mouse and is directed against the human monoclonal Ab<br />

(mAb) 2F5, which broadly and potently neutralizes primary HIV-1 isolates<br />

[3]. Ab2/3H6 which has been characterized previously [4,5] is able to mimic<br />

Page 92 of 181<br />

the antigen recognition site of 2F5 and therefore it is suggested as a<br />

putative candidate for an HIV-1 vaccine.<br />

We investigated the potential of Ab2/3H6 by immunization of Fab<br />

fragments and fusion proteins with interleukin 15 (IL15) and tetanus toxin<br />

(TT) tags as immune modulators. After three prime/boost administrations<br />

rabbit sera were purified and analyzed for 2F5-like specific Abs. Further,<br />

the 2F5-like Abs from the sera were enriched by affinity purification and<br />

characterized for their binding affinity to 2F5.<br />

In an additional approach we applied different humanization methods to<br />

reduce the immunogenicity of the originally mouse derived Ab2/3H6. The<br />

mouse variable regions of Ab2/3H6 were subjected to three different<br />

humanization methods, namely resurfacing, CDR-grafting and superhumanization.<br />

Four differently humanized Ab2/3H6 variants were<br />

characterized for their binding affinity to 2F5 in comparison to the original<br />

Ab2/3H6.<br />

Results: To evaluate the humoral immune response of Ab2/3H6 we<br />

designed Ab2/3H6 Fab fusion proteins with IL15 and TT. Recombinant CHO<br />

cell lines were established and after protein purification New Zealand white<br />

rabbits were immunized with the Ab2/3H6 Fab variants. Ten days after the<br />

final boost sera were collected and analyzed for total rabbit IgG levels. After<br />

proteinA affinity purification of the sera the isolated rabbit IgGs were tested<br />

for Ab2/3H6 Fab and recombinant gp140 (UG37) specificity (Figure 1A).<br />

Further an affinity enrichment step using a UG37/ELDKWA column was<br />

performed and the obtained Ab3 fraction was tested on UG37 (Figure 1B)<br />

and additionally on the original 2F5 epitope ELDKWA (Figure 1C). Finally the<br />

Ab3 fraction was tested for binding affinity to the UG37 in a bio-layer<br />

interferometry assay which showed that the Ab3 fraction has a 6.6 fold<br />

reduced affinity towards UG37 compared to the mAb 2F5 (Table 1).<br />

For the humanization approach three different methods were chosen. The<br />

“resurfaced” variant (RS3H6) was developed by a computer model and<br />

surface exposed amino acids in the murine framework (FR) were substituted<br />

by residues usually found at equivalent positions in human Abs. The<br />

“superhumanized” form (SH3H6) was designed by structural homologies<br />

between the murine Ab2/3H6 CDRs and human germline CDRs. The most<br />

homologous human germline Ab was then used as acceptor FR. For the<br />

“CDR-grafted” variant two versions were expressed. An “aggressive” graft<br />

(GA3H6) harbouring less backmutations, making the grafted Ab more<br />

human-like and a “conservative” graft (GC3H6) with more backmutations.<br />

The different “reshaped” variable regions of Ab2/3H6 were expressed in<br />

CHO-cells as IgG1 molecules. The obtained “humanized” Ab variants were<br />

further characterized by competition ELISA (Figure 1D).<br />

RS3H6 and GC3H6 showed a nearly identical slope of a concentration<br />

dependent 2F5 inhibition compared to the chimeric (ch) 3H6. RS3H6, GC3H6<br />

or ch3H6 complexed 50% of mAb 2F5 with a five fold molar excess. GA3H6<br />

was able to bind 50% of 2F5 in a 15 fold excess. In contrast, SH3H6 did not<br />

interact with 2F5 even in an 80 fold excess which is comparable to the


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P64) (A) Binding of purified rabbit IgGs immunized with different Fab preparations and control (pre-immun IgG fraction) to Ab2/<br />

3H6Fab and recombinant HIV-1 gp140 (UG37); (B) Binding of the Ab3 (2F5-like) Ab fraction purified from pooled rabbit IgG fractions to recombinant HIV-<br />

1 gp140 (UG37) and (C) synthetic 2F5 epitope (ELDKWA); (D) Competition ELISA. Biotinylated 2F5 (2F5-B) was allowed to complex with serial dilutions of<br />

Ab2/3H6 mutants and afterwards uncoupled 2F5-B was determined on an epitope pre-coated ELISA plate. The optical density resulting from binding of<br />

2F5-B to the synthetic epitope GGGELDKWASL is plotted versus the logarithm of the concentration of Ab2/3H6 mutants.<br />

negative control (unspecific IgG). The ELISA results were supported by<br />

binding affinity experiments with 2F5 in a bio-layer interferometry assay:<br />

RS3H6, GC3H6 and ch3H6 have similar binding affinity to 2F5 whereas<br />

GA3H6 has a 2-fold reduction in affinity.<br />

Discussion and conclusion: The Ab2/3H6 was generated to be used as<br />

an HIV-1 vaccine, based on the induction of 2F5-like Abs (Ab3s). First we<br />

did a “proof of concept” andimmunizedrabbitswithdifferentAb2/<br />

3H6Fab fusion proteins to induce 2F5-like Abs. Immunochemical analyses<br />

showed that the use of IL15 and TT as immune modulators do not<br />

Page 93 of 181<br />

significantly enhance total rabbit IgG levels or the production of Ab2/<br />

3H6Fab specific Abs. However it could be demonstrated that UG37<br />

binding Abs (Ab3s) were induced significantly (Figure 1A). Quantification<br />

of various purification steps revealed that only 0.7% of the rabbit IgG<br />

fraction contained UG37/ELDKWA binding Abs. This affinity enriched Ab3<br />

fraction shows significant binding to UG37 and reduced binding to the<br />

synthetic 2F5 epitope ELDKWA in ELISA (Figure 1B/C). Affinity binding<br />

studies showed that the obtained Ab3 fraction has only a 6.6-fold lower<br />

affinity to the recombinant UG37 protein compared to 2F5 (Table 1).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P64) Summary of k-values for binding to UG37<br />

Samples [c=400nM] kon [1/Ms] koff [1/s] KD [nM]<br />

mAb 2F5 1.66 x10 04<br />

3.09 x10 -04<br />

18.65<br />

Ab3 fraction 1.56 x10 04<br />

1.91 x10 -03<br />

122.90<br />

Control IgG fraction n.d. n.d. n.d.<br />

In a next step different humanization methods were evaluated. The<br />

“resurfaced” and the “conservative” grafted variants showed similar binding<br />

properties to 2F5 as the original Ab2/3H6 version, while the “aggressively”<br />

graftedAbhada2-foldloweraffinityandthe“superhumanized” form<br />

completely lost the ability to bind to 2F5.<br />

We postulate that after humanizing the framework regions of Ab2/3H6 the<br />

immune response will be focused towards the paratope of the Ab2 and<br />

therefore enhance elicitation of Ab3 when administered during human<br />

therapy. Therefore affinity to 2F5 is not the crucial factor in evaluating the<br />

best candidate for a clinical study. Our results show that the Ab2/3H6<br />

variants RS3H6 and GA3H6 would be suitable candidates for future studies.<br />

Acknowledgement: This work was funded by Austrian Science Fund,<br />

Vienna (P20603-B13)<br />

References<br />

1. Jerne N: Towards a network theory of the immune system. Ann Immunol<br />

(Paris) 1974, 125C:373-389.<br />

2. Kunert R, Weik R, Ferko B, Stiegler G, Katinger H: Anti-idiotypic antibody<br />

Ab2/3H6 mimics the epitope of the neutralizing anti-HIV-1 monoclonal<br />

antibody 2F5. AIDS 2002, 16:667-668.<br />

3. Wolbank S, Kunert R, Stiegler G, Katinger H: Characterization of human<br />

class-switched polymeric (immunoglobulin M [IgM] and IgA) anti-human<br />

immunodeficiency virus type 1 antibodies 2F5 and 2G12. J Virol 2003,<br />

77:4095-4103.<br />

4. Gach J, Quendler H, Weik R, Katinger H, Kunert R: Partial humanization and<br />

characterization of an anti-idiotypic antibody against monoclonal<br />

antibody 2F5, a potential HIV vaccine? AIDS Res Hum Retroviruses 2007,<br />

23:1405-1415.<br />

5. Gach J, Quendler H, Strobach S, Katinger H, Kunert R: Structural analysis<br />

and in vivo administration of an anti-idiotypic antibody against mAb<br />

2F5. Mol Immunol 2008, 45:1027-1034.<br />

Figure 1(abstract P65) Cell growth and cell viability.<br />

Page 94 of 181<br />

P65<br />

Application of hydrocyclones for continuous cultivation of SP-2/0 cells<br />

in perfusion bioreactors: Effect of hydrocyclone operating pressure<br />

Elsayed A Elsayed 1,2* , Roland Wagner 3<br />

1 Advanced Chair for Proteomics & Cytomics Research, Zoology Department,<br />

Faculty of Science, King Saud University, 11451 Riyadh, Kingdom of Saudi<br />

Arabia; 2 Natural & Microbial Products Dept., National Research Centre, Dokki,<br />

Cairo, Egypt; 3 Technology Development Department, Rentschler<br />

Biotechnology GmbH, D-88471 Laupheim, Germany<br />

E-mail: eaelsayed@ksu.edu.sa<br />

BMC Proceedings 2011, 5(Suppl 8):P65<br />

Background: Hydrocyclones (HCs) have been recently extensively evaluated<br />

for their application in the separation of mammalian cells in perfusion<br />

bioreactors. The high centrifugal force derived within hydrocyclones is the<br />

key mechanism for cell separation inside such small-sized simple devices.<br />

This is usually accompanied by a very short residence time of the cells inside<br />

the separating equipment, usually not more than 0.2 s. Moreover, they have<br />

other specific characteristics, which highly recommend their application for<br />

the production of pharmaceutical products in perfusion cultivation<br />

bioreactors. They are characterized by their high performance, robustness,<br />

lack of movable parts, ease of in situ sterilization and suitability for cleaningin-place<br />

processes.<br />

Materials and methods: The hydrocyclone HC 2520, specially designed for<br />

cell cultivation, was used for separation of the recombinant mouse lymphoid<br />

cell line SP-2/0. It has a 25-mm underflow and 20-mm overflow orifice.<br />

A pulsation-free pumphead (Watson Marlow 505L) was fitted to a WM 505DU<br />

pump and the perfusion was performed intermittently. Cell were cultured on<br />

serum-free ZKT-1 medium supplemented with 5% foetal bovine serum.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 2(abstract P65) Hydrocyclone separation efficiency.<br />

Results: Effect of hydrocyclone operating pressure on cell growth<br />

and cell viability: The results obtained (Figure 1) showed that cells<br />

required an adaptation phase to adapt themselves to the HC operating<br />

pressure. Moreover, the length of the adaptation phase depends on the<br />

HC pressure. Generally, cells were able to grow with high viability after the<br />

operation of hydrocyclone at both tested pressure values (0.85 and 1.30<br />

bar). Maximal cell concentrations of about 8.2 × 10 6 and 6.0 × 10 6 mL -1<br />

were achieved at an operating pressure of 0.85 and 1.30 bar with a<br />

viability range from 92 to 98%, respectively. This corresponds to a 5- and<br />

3.5-fold increase than the batch cultivation, respectively.<br />

Effect of hydrocyclone operating pressure on separation efficiency<br />

and separated cell quality: Results (Figure 2) showed that increasing<br />

operating pressure from 0.85 to 1.30 bar increased the average total<br />

separation efficiency from 89 to 95%. Additionally, results also showed that<br />

hydrocyclone preferably separates more viable cells in the underflow, and<br />

hence, back to the bioreactor system. Thus, more dead cells are separated<br />

in the overflow leaving the bioreactor system, which will improve system<br />

viability and product quality.<br />

Conclusions: - Hydrocyclone can be successfully used for cell separation in<br />

continuous mammalian perfusion bioreactors.<br />

- Cells adapt themselves to the shear forces inside the HC without being<br />

adversely affected by the pressure.<br />

- Higher separation efficiencies up to 96% can be achieved depending on<br />

the operating HC pressure.<br />

- HCs separate preferably more living cells in the underflow, thus more<br />

dead cells are leaving the system, which will finally lead to the<br />

improvement of system viability and product quality.<br />

P66<br />

Improving volumetric productivity of a stable human CAP cell line by<br />

bioprocess optimization<br />

Ruth Essers * , Helmut Kewes, Gudrun Schiedner<br />

CEVEC Pharmaceuticals GmbH, Gottfried-Hagen-Str. 62, D-51105 Köln,<br />

Germany<br />

E-mail: essers@cevec.com<br />

BMC Proceedings 2011, 5(Suppl 8):P66<br />

Page 95 of 181<br />

Background: For the production of recombinant proteins, a human cellderived<br />

expression technology can offer significant advantages with<br />

respect to protein quality, serum half-life and safety. High volumetric<br />

productivity with first-class quality is the ultimate ambition during<br />

process development.<br />

CEVEC’s proprietary expression system based on human amniocytes offers<br />

significant advantages for the production of complex human proteins<br />

and antibodies. The key benefits are the stable and high expression of<br />

recombinant proteins with human type posttranslational modifications, the<br />

robust growth behaviour with competitive high cell densities and the easy<br />

handling in serum free suspension. CAP cells meet all regulatory<br />

requirements, they are of non-tumour origin and from an ethically accepted<br />

source.<br />

In order to test the performance of CAP cells for the production of very<br />

complex proteins, stable C1-inhibitor expressing CAP cells were developed.<br />

C1-inhibitor is a serine protease inhibitor (serpin) and one of the most<br />

heavily glycosylated plasma proteins bearing numerous complex N- and Oglycans.<br />

Material and methods: Cells: CAP cells stably expressing C1-inhibitor<br />

Base medium: Protein Expression medium PEM (Gibco #12661013)<br />

containing 4mmol*L -1 glutamine (Invitrogen #25030024)<br />

Supplements: Soy Peptone E110 (OrganoTechnie #AI885), Glucose (Sigma<br />

#G8679), Valproic acid VPA (Sigma #4543)<br />

ELISA: In-house C1-inhibitor ELISA using serum derived C1-inhibitor as<br />

standard<br />

SDS-PAGE/Western Blot: In-house Western Blot for C1-inhibitor<br />

To investigate the influence of different hydrolysates and supplements<br />

under controlled conditions we use DASGIP´s parallel bioreactor system for<br />

cell culture. See table 1 for standard physical process parameters.<br />

Results: Parental CAP cells were transfected with a plasmid containing<br />

expression cassettes for the human C1-inhibitor driven by the CMV<br />

promoter, and a Blasticidine resistancegeneforselection.Outofthree<br />

stable pools, one pool was selected for further single cell cloning by<br />

limiting dilution. From initial 251 single cell clones, the best 5 were<br />

chosen for further scale-up. Subsequent process development was<br />

carried out with one clone showing the best performance in growth<br />

and expression.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P66) Standard physical process<br />

parameters for DASGIP fermentation<br />

process parameter<br />

T 37°C<br />

V 0.6L<br />

stirring marine impeller, 120rpm<br />

aeration sparging, 0.03vvm<br />

pH 7.0 (CO2/ 0.5M NaOH)<br />

DO 40% (air saturation)<br />

starting cell density 3-5E5mL -1<br />

First we tested the influence of different media supplements shown<br />

before to improve cell growth and productivity in CAP cells. Either<br />

additional glucose, glutamine, pyruvate, soy peptone, tryptone plus,<br />

tryptone or an in-house mixture of R3-IGF, transferrin, SyntheChol and<br />

progesterone were added to the base medium. Cells were cultivated in<br />

shake flasks and cell densities, viabilities, metabolites and product<br />

concentration were monitored continuously.<br />

Whereas the in-house mixture lead to higher growth rates, the product<br />

yields increased upon the addition of glucose (1.2-fold), soy peptone or<br />

tryptone (1.5-fold).<br />

Based on these results, we tested additional hydrolysates from cotton seed,<br />

wheat gluten, rice protein and soy. Hydrolysates and extra glucose were<br />

added to the initial medium and cells were cultivated in shake flasks.<br />

Only medium supplemented with soy peptone II (up to 1.7-fold) and with<br />

cotton seed hydrolysate (up to 2.4-fold) showed better performance as<br />

compared to the control with extra glucose but without hydrolysate.<br />

In addition to medium supplementation we determined the optimal pH<br />

value for good growth rates, high product quantity and quality. The<br />

different batches (pH unregulated, pH 7.0, 7.2 or 7.4) did not differ in<br />

maximal product concentrations but in product qualities. This was<br />

monitored with SDS-PAGE and western blot. Due to additional specific<br />

bands at pH7.2 and pH7.4 either caused by degradation or incomplete<br />

glycosylation we decided to continue with pH7.0 for following process<br />

development steps.<br />

Under controlled conditions additional glucose alone or either with soy<br />

peptone I, soy peptone II or cotton seed hydrolysate were supplemented to<br />

the base medium. Parallel fermentations with different enriched media were<br />

started at pH7.0 and cell densities of 3.0*10 5 mL -1 under controlled<br />

conditions. Cotton seed hydrolysate increased growth, but highest cell<br />

density could be observed with soy peptone II or without hydrolysate<br />

addition. The maximum product concentration was higher compared to the<br />

control in all peptone-fed fermentations. Supplementing soy peptone II or<br />

cotton seed hydrolysate almost doubled maximum product concentration.<br />

As a next step, we examined the effect of valproic acid (VPA) on productivity.<br />

VPA is a short-chain fatty acid used as well established drug and classified<br />

histone deacetylase inhibitor. We set up two cultures with glucose and soy<br />

peptone II and two cultures with glucose and cotton seed hydrolysate. In<br />

each case, one of the duplicate cultures was additionally supplemented with<br />

4mmol*L -1 VPA.<br />

Initial cell densities were 3.5*10 5 mL -1 in all four parallel controlled cultures.<br />

The addition of VPA was carried out at viable cell densities of 3.0*10 6 mL -1 .<br />

Addition of VPA resulted in 1.6-2.0-fold increase in productivity compared<br />

to the corresponding control cultures without VPA<br />

To determine the product quality we performed Western blots with samples<br />

from the supernatant of the batches supplemented with VPA. The highest<br />

protein quality was obtained from the Soy peptone II-supplemented<br />

cultures.<br />

We confirmed the results with 8 (parallel) runs and observed consistent<br />

results for cell densities, growth behaviour, product titers, cell specific and<br />

volumetric productivities.<br />

Conclusions: The optimized fed batch strategy starting with 2g*L -1 extra<br />

glucose and 4g*L -1 soy peptone II and addition of 4mmol*L -1 VPA at a<br />

viable cell denstity of 3.5*10 6 mL -1 yields 4.3-fold higher volumetric<br />

productivity as compared to the batch culture (Fig.1). With the optimized<br />

fed batch we are able to produce 200-250 mg*L -1 C1-inhibitor.<br />

P67<br />

Development of a triculture based system for improved benefit/risk<br />

assessment in pharmacology and human food<br />

Alexandra Bazes, Géraldine Nollevaux, Régis Coco, Aurélie Joly,<br />

Thérèse Sergent, Yves-Jacques Schneider *<br />

Biochimie cellulaire, nutritionnelle & toxicologique, Institut des Sciences de la<br />

Vie & UCLouvain, Croix du Sud 5/3, 1348 Louvain-la-Neuve, Belgium<br />

E-mail: yjs@uclouvain.be<br />

BMC Proceedings 2011, 5(Suppl 8):P67<br />

Caco-2 cells are widely used for both mechanistic studies in molecular cell<br />

biology as well as for studies aiming at estimating, in vitro, the<br />

bioavailability of drug candidates, xenobiotics, food compounds …. This<br />

Figure 1(abstract P66) Process development C1-inhibitor expressing CAP cell line – Improvement of volumetric productivity.<br />

Page 96 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P67) In vitro models characterisation: Cell monolayer integrity was determined by LY transport. LY<br />

was incubated with cell monolayers for 180 min at 37°C (n=3) and it was quantified by fluorimetry. The results were<br />

expressed as % of total fluorescent<br />

Caco-2 HT29-MTX Caco-2/HT29-MTX Caco-2/Raji Caco-2/HT29-MTX/Raji<br />

TEER (Ω.cm 2 ) 283±70 122±31 122±19 88±27 60±17<br />

Lucifer Yellow transport (% fluorescence) 0,40±0,02 6,42±0,09 3,25±0,06 7,16±0,23 5,07±0,22<br />

results largely from the spontaneous differentiation of this human colon<br />

adenocarcinoma line into enterocytes-like cells in classical culture<br />

conditions, as well as from the use of bicameral culture inserts including<br />

porosity calibrated filters.<br />

Although Caco-2 cells represent the current “golden standard” of in vitro<br />

models of the human intestinal barrier, there is a trend to develop new<br />

systems mimicking more closely the intestinal epithelium for pharmaceutical<br />

and toxicological studies. In particular, goblet cells may bring a main<br />

constituent of the intestinal barrier as secreting mucus and contribute to the<br />

permeability met in vivo. In addition, gut associated M cells offer a putative<br />

way for oral delivery of nanoencapsulated therapeutic peptides and mucosal<br />

vaccines.<br />

Material and methods: The triculture model was adapted by combining<br />

the in vitro model of the human follicle-associated epithelium (including Mlike<br />

cells) according to the protocol of des Rieux et al. (2007) and the coculture<br />

of Caco-2 and HT29-5M1 cells from Nollevaux et al. (2006). Briefly,<br />

Caco-2 cells with or without HT29-5M1 cells were seeded on 12-wells<br />

Transwell inserts (3.0 μm pore diameter, Costar, Elscolab, Kruibeke, BE) and<br />

cultivated for 3 days in DMEM supplemented with 10% foetal calf serum<br />

Page 97 of 181<br />

(FCS). Some inserts were then inverted and a piece of silicon rubber (Labo-<br />

Modern, Queveaucamps, BE) placed around the basolateral side. Then,<br />

inserts were transferred into a Petri dish pre-filled with culture medium and<br />

maintained for 10 days with the basolateral medium refreshed every 2 days.<br />

Raji-B cells, suspended in DMEM + 10% FCS, were added to the basolateral<br />

compartment of the inserts. The tricultures were maintained for 5 days. Cocultures<br />

were cultured under the same conditions, but without the addition<br />

of Raji-B cells or HT29-5M1 cells.<br />

Cell monolayer integrity, both in mono-, co- and tri-cultures, was controlled<br />

by measurement of the transepithelial electrical resistance (TEER, Millipore<br />

Millicell® ERS, Billerica, MA) and by evaluation of cell permeability to Lucifer<br />

Yellow (LY; 457 Da, Sigma-Aldrich, St-Louis, MO).<br />

After integrity experiments, cells were washed in PBS, fixed with acetone<br />

and permeabilised with 1% (v/v) Triton X-100. Cells were then washed with<br />

PBS, blocked with 1% BSA and washed again with PBS, and then incubated<br />

with mouse anti-MUC5AC at 1:100 dilution, rinsed in PBS and incubated<br />

with Alexa Fluor 488 goat anti-mouse 10µg/ml and Rhodamine-Phalloidin<br />

4U/ml (Invitrogen, Eugene, OR). Finally, cells were mounted with Ultracruz<br />

Mounting Medium (Santa Cruz Biotechnology, Santa Cruz, CA). The<br />

Figure 1(abstract P67) Immunofluorescent localization of different cell types. The nuclei of cells were stained in blue with the mounting medium.<br />

Enterocyte actin was stained with rhodamine-phalloidin (red) and goblet-like cells were MUC5AC-labelled (green). The M-like cells were detected by their<br />

lack of microvilli.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

monolayers were viewed using a Zeiss LSM 710 confocal laser scanning<br />

microscope (Carl Zeiss MicroImaging GmbH, Jena, DE).<br />

Results and discussion: To validate the triculture system as an intestinal<br />

barrier model, transport studies were used to determine the culture<br />

integrity and the permeability capacity with TEER measurement and<br />

assessment of the passage of LY. Furthermore, a morphological analysis<br />

was initiated to evaluate the proportion of the M cells, goblet cells and<br />

enterocytes and their implication in the intestinal epithelium model.<br />

Cell monolayer integrity and permeability properties: Adecreaseof<br />

TEER was observed in the different co-culture systems. This should, most<br />

probably, be correlated to an increase of the barrier permeability, upon<br />

addition of HT29-MTX or/and Raji cells in the system (Table 1).<br />

The effect of the addition of goblet-like cells or of the conversionof<br />

Caco-2 cells into M-like cells suggests a role of these cells in the ability of<br />

the intestinal cells to increase their absorbtion of hydrophilic molecules,<br />

as observed by the increase of the permeability of LY in the co- and triculture<br />

systems in comparison with the control.<br />

The contribution of the different cell types in the triculture system further<br />

allows the integration of mechanisms such as transcytosis (M cells), the<br />

presence of mucus (goblet-like cells), and the effectiveness of the tight<br />

junctions in the culture system, as compared to a standard Caco-2 model.<br />

Localisation of the intestinal cell types: The intestinal cell types were<br />

detected by fluorescent labelling and confocal analysis (Figure 1). Actin<br />

(red) and MUC5AC (green) were restricted respectively on enterocyte and<br />

goblet-like cells. The M-like cells appeared within the enterocyte layer<br />

and were identified by their lack of microvilli (actin) at their apical<br />

surface. The mucus produced by goblet-like cells was on the surface<br />

(MUC5AC). In the triculture system, there was close contact between<br />

HT29-MTX, Raji and Caco-2 cells but the goblet-like cells have tendency<br />

to cluster together.<br />

Conclusion, perspectives: These results clearly indicate that the<br />

cocultivation of 3 different cell types allows to reconstitute a cell culture<br />

system that should better mimic the intestinal barrier, specially to<br />

investigate its interaction with nanoparticles. This should facilitate a<br />

better evaluation of pharmacological or toxicological properties of these<br />

materials in food and drugs.<br />

We are now developing additional co-culture systems involving f.i.<br />

RAW264.7 macrophages to further determine the interactions involved in<br />

inflammatory processes; HepG2 cells to estimate the intestinal and<br />

hepatic effects in presystemic biotransformations.<br />

References<br />

1. des Rieux A, Fievez V, Théate I, Mast J, Préat V, Schneider YJ: An improved<br />

in vitro model of human intestinal follicle-associated epithelium to<br />

study nanoparticle transport by M cells. Eur J Pharm Sci 2007,<br />

30(5):380-391.<br />

2. Nollevaux G, Deville C, El Moualij B, Zorzi W, Deloyer P, Schneider YJ,<br />

Peulen O, Dandrifosse G: Development of a serum-free co-culture of<br />

human intestinal epithelium cell-lines (Caco-2/HT29-5M21). BMC Cell Biol<br />

2006, 2:7-20.<br />

P68<br />

Daphnane diterpene hirsein B downregulates melanogenesis in B16<br />

murine melanoma cells by cAMP pathway inhibition<br />

Myra O Villareal 1 , Junkyu Han 1,2 , Kenjiro Ikuta 3 , Hiroko Isoda 1,2*<br />

1 Graduate School of Life and Environmental Sciences, University of Tsukuba,<br />

Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan; 2 Alliance for Research on<br />

North Africa (ARENA), University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki,<br />

305-8572, Japan; 3 Yokohama Corporate Research Laboratories, Mitsubishi<br />

Rayon Co., Ltd, Tsurumi-ku, Yokohama, Kanagawa, 230-0053, Japan<br />

E-mail: isoda.hiroko.ga@u.tsukuba.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):P68<br />

Background: Skin pigmentation serves as protection against ultravioletinduced<br />

skin damage through melanin’s optical and chemical filtering<br />

properties [1]. Although melanin plays and important role in skin protection,<br />

excessive melanin production or hyperpigmentation may lead to skin<br />

cancer. Recently, the inhibition of melanogenesis has been considered as a<br />

valid therapeutic target for the management of advanced melanotic<br />

melanomas [2] which increases the need for melanogenesis inhibitors<br />

that are of plant origin and are not cytotoxic to mammalian cells. The<br />

biosynthesis of the pigment melanin is catalyzed by the melanogenic<br />

Table 1(abstract P68) Differentially-expressed genes in<br />

hirsein B-treated B16 murine melanoma cells as<br />

determined by DNA microarray<br />

Biological Process Differentially<br />

expressed genes<br />

Up- or Downregulated<br />

Melanin biosynthesis Mitf, Mc1r Downregulated<br />

Melanosome transport Rab27a, Mlph,<br />

Myo5A, Myo7A<br />

Downregulated<br />

Negative regulation of<br />

transcription from RNA<br />

polymerase II promoter<br />

Sorbs3 Downregulated<br />

Wnt signaling pathway Ppap2b, Wisp,<br />

Kremen1<br />

Upregulated<br />

Cell cycle regulation Gadd45b, Csnkl Upregulated<br />

Activation of MAPK signaling<br />

pathway<br />

Gadd45b , Pxn,<br />

Map2k3, Met, Avpi1,<br />

Spag9<br />

Page 98 of 181<br />

Upregulated<br />

Cytoskeleton organization Pxn Upregulated<br />

Protein phosphorylation Mapkapk3 Upregulated<br />

enzymes tyrosinase, tyrosinase related protein 1 and the dopachrome<br />

tautomerase, the transcriptional regulation of which is being regulated by<br />

the microphthalmia associated transcription factor (Mitf) [3]. Previously, we<br />

have reported that hirsein B (HB) or 5b-hydroxyresiniferonol-6a,7a-epoxy-<br />

12b-coumaroyloxy-9,13,14-ortho-decanoate from Thymelaea hirsuta [4] has<br />

antimelanogenesis effect (without cytotoxicity) on B16 murine melanoma<br />

cells by downregulating the expressions of the Mitf gene and the<br />

melanogenic enzymes’ genes [5]. The exact mechanism by which hirsein B<br />

inhibited the Mitf gene expression, however, has not yet been determined.<br />

In melanogenesis, the Mitf gene expression can be regulated through the<br />

cAMP pathway or the Wnt signaling pathway. This study aimed to<br />

determine the mechanism underlying the inhibitory effect of HB on Mitf<br />

gene in B16 murine melanoma cells.<br />

Materials and methods: Total RNA was isolated from B16 murine<br />

melanoma cells (Riken Cell Bank, Tsukuba, Japan) and used for DNA<br />

microarray analysis, using chips of 528 spots loaded with 265 genes prepared<br />

by Genopal (Mitsubishi Rayon Co., Ltd, Tokyo, Japan), to determine the<br />

expressions of genes for melanogenesis, membrane-bound receptors,<br />

tyrosine kinase regulation, melanosome transport, and other cell signal<br />

regulation-related genes (including the housekeeping and negative control<br />

genes). To validate the results, real-time PCR, using TaqMan FAST 7500<br />

(Applied Biosystems, Foster City, CA, USA) and specific TaqMan primers<br />

(Applied Biosystems, Foster City, CA, USA) for the differentially-expressed<br />

genes, was performed.<br />

Results: Results showed that the expressions of the Mitf gene and the<br />

melanogenic enzymes’ genes were downregulated, verifying our previous<br />

report [5]. In addition, the expression of the gene for melanocortin 1<br />

receptor (Mc1r) of the cAMP pathway was downregulated while most of<br />

the genes that were upregulated are those involved in the Wnt signaling<br />

pathway (Table 1).<br />

In mouse, peptide hormones from the pituitary gland bind to the MC1R<br />

and stimulate melanin production through the cAMP/PKA signalling<br />

pathway [6], by inducing changes in the protein phosphorylation and<br />

gene expression, through the MITF gene product.<br />

Conclusions: The results obtained suggest that the significant<br />

antimelanogenesis effect of hirsein B is through the inhibition of the<br />

expression of the Mc1r gene of the cAMP pathway. HB may therefore be<br />

used as a treatment for hyperpigmentation due to its significant<br />

melanogenesis downregulation effects in B16 cells or as a pretreatment<br />

for melanotic melanomas.<br />

Acknowledgment: This study was partially supported by the Science and<br />

Technology Research Partnership for Sustainable Development (SATREPS).<br />

References<br />

1. Ahene AB, Saxena S, Nacht S: Photoprotection of solubilized and<br />

microdispersed melanin particles. Melanin, Its Role in Human<br />

Photoprotection Overland Park, KS: Valendmar: Zeise L, Chedekel M,<br />

Fitzpatrick TB 1995, 255-269.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

2. Slominski A, Zbytek B, Slominski R: Inhibitors of melanogenesis increase<br />

toxicity of cyclophosphamide and lymphocytes against melanoma cells.<br />

Int J Cancer 2009, 124:1470-1477.<br />

3. Slominski A, Tobin DJ, Shibahara S, Wortsman J: Melanin pigmentation in<br />

mammalian skin and its hormonal regulation. Physiol Rev 2004,<br />

84:1155-1228.<br />

4. Miyamae Y, Orlina-Villareal M, Isoda H, Shigemori H: Hirseins A and B,<br />

daphnane diterpenoids from Thymelaea hirsuta that inhibit<br />

melanogenesis in B16 melanoma cells. J Nat Prod 2009, 72:938-941.<br />

5. Villareal M, Han J, Yamada P, Shigemori H, Isoda H: Hirseins inhibit<br />

melanogenesis by regulating the gene expressions of Mitf and<br />

melanogenesis enzymes. Exp Dermatol 2010, 19:617-627.<br />

6. Busca R, Ballotti R: Cyclic AMP a key messenger in the regulation of skin<br />

pigmentation. Pigment Cell Res 2000, 13:60-69.<br />

P69<br />

The neuroprotective effects of electrolyzed reduced water and its<br />

model water containing molecular hydrogen and Pt nanoparticles<br />

Hanxu Yan 1 , Taichi Kashiwaki 2 , Takeki Hamasaki 2 , Tomoya Kinjo 1 ,<br />

Kiichiro Teruya 1,2 , Shigeru Kabayama 3 , Sanetaka Shirahata 1,2*<br />

1 Graduate School of Systems Life Sciences, Kyushu University, 6-10-1<br />

Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan; 2 Department of Bioscience<br />

and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 812-<br />

8581, Japan; 3 Nihon Trim Co. Ltd., 1-8-34 Oyodonaka, Kita-ku, Osaka 531-<br />

0076, Japan<br />

E-mail: sirahata@grt.kyushu-u.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):P69<br />

Page 99 of 181<br />

Background: Human brain is the biggest energy consuming tissue in<br />

human body. Although it only represents 2% of the body weight, it<br />

receives 20% of total body oxygen consumption and 25% of total body<br />

glucose utilization. For that reason, brain is considered to be the most<br />

vulnerable part of human body against the reactive oxygen species (ROS),<br />

a by-product of aerobic respiration. Oxidative stress is directly related to a<br />

series of brain dysfunctional disease such as Alzheimer’s disease,<br />

Parkinson’s disease etc. Electrolyzed reduced water (ERW) is a functional<br />

drinking water containing a lot of molecular hydrogen and a small amount<br />

of platinum nanoparticles (Pt NPs, Table 1). ERW is known to scavenge ROS<br />

and protect DNA from oxidative damage [1]. We previously showed that<br />

ERW was capable of extending lifespan of Caenorhabditis elegans by<br />

scavenging ROS [2]. Molecular hydrogen could scavenge ROS and<br />

protected brain from oxidative stress [3]. Pt NPs are also a new type of<br />

multi-functional ROS scavenger [4].<br />

Materials and methods: In this research, we used TI-200S ERW derived<br />

from 2 mM NaOH solution produced by a batch type electrolysis device<br />

and model waters containing molecular hydrogen and synthetic Pt NPs of<br />

2-3 nm sizes as research models of ERW to examine the anti-oxidant<br />

capabilities of ERW on several kinds of neural cells such as PC12, N1E115,<br />

and serum free mouse embryo (SFME) cells. We pretreated the ERW and<br />

200 μM H2O2 and examined the neuroprotective effects of ERW on PC12,<br />

N1E115 and SFME cells, using WST-8 method. We also examined the<br />

intracellular ROS scavenging effects of ERW on N1E115 cells after<br />

pretreated cells with ERW and H 2O 2 using DCFH-DA. We checked the<br />

protective effects of ERW on mitochondria and cytoplasm by Rh123 and<br />

Fuo-3 AM stain. We also examined the ATP production of SFME cells after<br />

pretreated with ERW and H 2O 2 by Bioluminescence Assay Kit. Finally, we<br />

Table 1(abstract P69) Characteristics of the water samples.The characteristics of water samples were determined<br />

immediately after the preparation of ERW. ERW, electrolyzed reduced water; CW, activated charcoal-treated water.<br />

The pH values were shown as average ± standard deviation (N = 5). The values of DH, DO and Pt NPs were shown the<br />

minimum and maximum values after 5 independent measurements<br />

MQ (NaOH) TI-200 ERW TI-9000 CW TI-9000 ERW<br />

pH 11.3 ± 0.1 11.6 ± 0.1 7.9 ± 0.1 9.6 ± 0.2<br />

Dissolved Hydrogen (mM) 0 0.2– 0.45 0 0.1– 0.25<br />

Dissolved Oxygen (μM) 0 3.1– 78.1 0 0– 21.9<br />

Pt NPs (nM) 0 0.5– 12.8 0 0– 3.6<br />

Redox potential value (mV) + 350 -659 - -<br />

Figure 1(abstract P69) Protective effect of ERW on H 2O 2-induced neuroblastoma N1E115 cells death. Cells were treated with water samples (ERW<br />

and control ultrapure water with same pH with ERW) and 200 μM H 2O 2 for 24 h. Cell viabilities were assayed by WST-8 method. N=3, * p < 0.05.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

used dissolved hydrogen (DH) and Pt NPs as research models to examine<br />

their neuroprotective effects.<br />

Results: ERW significantly reduced the cell death induced by H 2O 2<br />

pretreatment (Figure 1). ERW also scavenged the intracellular ROS and<br />

prevented the decrease of mitochondrial membrane potential and ATP<br />

production induced by ROS. We also examined the neuroprotective<br />

effects of molecular hydrogen and Pt NPs and showed that both<br />

molecular hydrogen and Pt NPs contributed to the neuroprotective<br />

effects of ERW.<br />

Conclusion: The results suggest that ERW is beneficial for the prevention<br />

and alleviation of oxidative stress-induced human neurodegenerative<br />

diseases.<br />

References<br />

1. Shirahata S, Kabayama S, Nakano M, Miura T, Kusumoto K, Gotoh M,<br />

Hayashi H, Otsubo K, Morisawa S, Katakura Y: Electrolyzed-reduced water<br />

scavenges active oxygen species and protects DNA from oxidative<br />

damage. Biophys Biochem Res Commun 1997, 234:269-274.<br />

2. Yan H, Tian H, Kinjo T, Hamasaki T, Tomimatsu K, Nakamichi N, Teruya K,<br />

Kabayama S, Shirahata S: Extension of the lifespan of Caenorhabditis<br />

elegans by the use of electrolyzed reduced water. Biosci Biotech Biochem<br />

2010, 74:2011-2015.<br />

3. Ohsawa I, Ishikawa M, Takahashi K, Watanabe M, Nishimaki K, Yamagata K,<br />

Katsura K, Katayama Y, Asoh S, Ohta S: Hydrogen acts as a therapeutic<br />

antioxidant by selectively reducing cytotoxic oxygen radials. Nature Med<br />

2007, 13:688-694.<br />

4. Hamasaki T, Kashiwagi T, Imada T, Nakamichi N, Aramaki S, Toh K,<br />

Morisawa S, Shimakoshi H, Hisaeda Y, Shirahata S: Kinetic analysis of<br />

superoxide anion radical-scavenging and hydroxyl radical-scavenging<br />

activities of platinum nanoparticles. Langmuir 2008, 24:7354-7364.<br />

P70<br />

Development and fine-tuning of a scale down model for process<br />

characterization studies of a monoclonal antibody upstream production<br />

process<br />

Mareike Harmsen * , Jimmy Stofferis, Laetitia Malphettes<br />

Cell Culture Process Development Group, Biological Process Development,<br />

UCB Pharma S.A, Braine-l’Alleud, 1420, Belgium<br />

E-mail: Mareike.Harmsen@ucb.com<br />

BMC Proceedings 2011, 5(Suppl 8):P70<br />

Background: It has always been an objective of process development<br />

and more recently it has also become a regulatory expectation to build<br />

robustness into and demonstrate proper control of a manufacturing<br />

process, thus ensuring that the biological product meets consistently its<br />

quality attributes and specifications. This is achieved mainly through<br />

systematic process development and understanding. Once a process is<br />

locked and ahead of consistency runs at the intended commercial scale,<br />

process characterization studies (PCS hereafter) further contribute to the<br />

demonstration of process robustness and the justification of process<br />

Page 100 of 181<br />

control ranges. These studies characterize the relationship between<br />

process parameters and process performance as well as product quality<br />

attributes. For practical reasons PCS are performed in a scale down model<br />

(SDM hereafter) of the manufacturing process studied. Therefore, it is<br />

essential to establish a SDM that is representative of the commercial<br />

scale.<br />

Here we describe a road map for developing a scale down model of a cell<br />

culture process for recombinant protein production. The cell culture process<br />

modeled was a 12,000 L scale fed batch process producing a monoclonal<br />

antibody.<br />

Methods: In a first step, the SDM was defined such that volume- dependent<br />

parameters (medium and feeds volumes, working volumes) were scaled<br />

down linearly and volume-independent ones (temperature, dissolved<br />

oxygen, pH, cell age) were kept at the same set-points as in the commercial<br />

scale bioreactor. Some cell culture process parameters such as agitation and<br />

aeration are difficult to scale down linearly due to bioreactor and sparger<br />

geometry and differences in controller systems. Hence determination of<br />

these parameters was first done by engineering scale down considerations.<br />

These considerations determined a theoretical small scale model used for<br />

proof of concept experiments. Process performance in initial scale down<br />

runs was then compared to commercial scale production in terms of cell<br />

growth, key metabolites, product accumulation.<br />

Two main deliverables were defined for the SDM:<br />

1. The model should exhibit process performance comparable to the<br />

commercial scale process. This was necessary to study the effects of input<br />

parameter variations during PCS.<br />

A way of assessing process performance is by monitoring in-process<br />

parameters during the SDM runs and comparing them to commercial<br />

scale batch data. Here the following evaluation parameters were chosen:<br />

Viable cell density and viabilities<br />

Metabolism indicators (namely off-line pH, dCO 2, osmolality, glucose and<br />

lactate concentrations)<br />

Product titer<br />

2. The model should deliver material with product quality attributes<br />

comparable to the material produced at the corresponding manufacturing<br />

scale process step (post cell harvest). This was necessary to study the<br />

effects of parameter changes on upstream product quality.<br />

Here the evaluation parameters were based on selected product quality<br />

specifications for the commercial scale batches. Namely:<br />

Oligosaccharide profiles<br />

Monomer/aggregate/fragment proportions<br />

Monomer,- Heavy-and Light chain patterns<br />

Acidic peak group species proportions<br />

Results: Proof of concept bioreactor runs using the theoretical SDM showed<br />

good comparability between the model and the commercial scale regarding<br />

process performance; however it was observed that the dissolved CO 2<br />

values were lower than in commercial scale. We consider this to be a<br />

consequence of geometrical bioreactor differences. The liquid surface-tovolume<br />

ratio is significantly higher in small scale than it is in large scale.<br />

Furthermore in small scale bioreactors a higher maximum volumetric gas<br />

Table 1(abstract P70) Product quality attributes of purified harvest material<br />

Product quality attribute Commercial scale reference<br />

sample<br />

SDM verification run 1 SDM verification run 2 SDM verification run 3<br />

Oligosaccharide mapping Profile comparable to standard Profile comparable to Profile comparable to Profile comparable to<br />

standard<br />

standard<br />

standard<br />

Monomer [%] 99<br />

Purity by size exclusion HPLC<br />

100 99 99<br />

Aggregate [%] 1 0 0 1<br />

Fragment [%] 0 0<br />

Monomer pattern by capillary gel electrophoresis<br />

1 0<br />

Monomer [%] 97 97 97 97<br />

Light and heavy chain pattern by capillary gel electrophoresis<br />

Light chain + heavy chain<br />

[%]<br />

99 99<br />

Acidic species by cation exchange chromatography<br />

98 99<br />

Acidic species [%] 30 30 28 32


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

flow rate was employed to compensate for a reduced gas bubble residence<br />

time. These two main differences resulted in increased CO 2 stripping.<br />

An overlay flow containing 6% CO 2 was applied to the headspace of the<br />

2 L vessels to reduce the stripping (fine –tuning).<br />

After establishing the CO2 overlay as part of the aeration strategy, three SDM<br />

verification runs were carried out. Cell growth, cell metabolism and Mab<br />

titers were monitored throughout the cultures. Lab scale data resulting from<br />

daily sampling was mostly within the average ± 3 x standard deviation (SD)<br />

of the commercial scale data and therefore comparable.<br />

Scale down model harvest material was compared to harvest material from<br />

the commercial scale with regards to their product quality attributes. The<br />

clarified cell culture fluid from both scales was protein A purified in<br />

laboratory scale and product quality was assessed (Table 1).<br />

Conclusions: Good comparability regarding process performance of the<br />

production bioreactor between the SDM runs and large scale data was<br />

demonstrated. Growth data, product titer, pH, osmolality, glucose and<br />

lactate profiles all showed the same trends. The tested output parameters<br />

where mostly within ± 3 x SD of the average commercial scale data.<br />

Product quality attributes of scale down samples were comparable to<br />

commercial scale product quality as shownbycomparisonwithclarified<br />

cell culture fluid samples from commercial scale.<br />

The verification results demonstrate that the systematic scale down approach<br />

applied in this work provided a laboratory scale model that was representative<br />

of the commercial scale process in terms of process performance and<br />

product quality of the generated material and was therefore suitable to be<br />

used in process characterization studies.<br />

Acknowledgements: We would like to thank Robert McCombie and Rav<br />

Ganesh for protein purification and Simon Briggs and François Delcroix<br />

for product quality analysis.<br />

P71<br />

Insect cell lines and baculoviruses as effective biocontrol agents of forest<br />

pests<br />

Guido F Caputo * , Sardar S Sohi, Sharon V Hooey, Lawrence J Gringorten<br />

Natural Resources Canada, Great Lakes Forestry Centre, Sault Ste. Marie, ON<br />

P6A2E5 CANADA<br />

E-mail: gcaputo@nrcan.gc.ca<br />

BMC Proceedings 2011, 5(Suppl 8):P71<br />

Background: Canada is a nation of trees. Canadian forests cover 50% of<br />

Canada’s land area and represent 10% of world’s forestedarea.Canada<br />

exports more processed forest products than any other country. It is also a<br />

host to a variety of forest insects both native and non native that limit the<br />

economic, recreational and wildlife habitat use. Our research focuses on<br />

initiating insect cell lines, determining nutritional requirements of cells,<br />

and developing low cost media for large scale propagation of insect<br />

pathogenic viruses as ecologically sound alternatives to chemical<br />

pesticides. Cell lines are used for bioassay and strain selection of viruses<br />

and bacterial toxins and for production of foreign gene products using<br />

baculovirus- and entomopoxvirus expression vectors. They offer a cleaner,<br />

viable alternative to insect larvae for producing viral pesticides.<br />

Since 1969, over 150 continuous cell lines have been produced for forest<br />

insect pest research at GLFC. This collection represents one of largest single<br />

site repository of frozen insect cell lines in the world. Cell lines developed<br />

include tissues of the eastern spruce budworm (Choristoneura fumiferana),<br />

western spruce budworm (Choristoneura occidentalis), forest tent caterpillar<br />

(Malacosoma disstria), tobacco hornworm (Manduca sexta), white-marked<br />

tussock moth (Orgyia leucostigma), red-headed pine sawfly (Neodiprion<br />

lecontei), gypsy moth (Lymantria dispar), white pine weevel (Pissodes strobi),<br />

the tarnished plant bug (Lygus lineolaris) and the ash and privet borer<br />

(Tylonotus bimaculatus). These cell lines represent six tissues of origin, namely<br />

neonate larvae, pre-pupae, embryos, ovaries, hemocytes, and midgut and<br />

four Insect Orders namely Lepidoptera, Hymenoptera, Coleoptera and<br />

Hemiptera.<br />

Presently, cell lines developed at GLFC are being used by 41 researchers<br />

in 8 Canadian provinces, 42 researchers in 21 US States and 28<br />

researchers in 12 countries worldwide.<br />

Biological control: Although chemical pesticides are generally more<br />

reliable and effective control agents, they are not environmentally<br />

acceptable as their negative impact far out weigh their benefits. We have<br />

developed methodologies to duplicate the in vivo process of the insect<br />

by producing both baculovirus phenotypes, the occlusion derived virus<br />

Page 101 of 181<br />

(ODV) and the budded virus (BV) in vitro. During the in vivo infection<br />

process hemolymph (BV) is collected and cultures are infected producing<br />

both phenotypes. BV, released in the media is used to infect other cells;<br />

ODV produced in the nuclei to infect other insects.<br />

Lawn assay: Trees produce numerous compounds with toxic and growth<br />

regulating properties to protect themselves against insect attack. We have<br />

developed a rapid in vitro agarose lawn assay for testing the toxicity of<br />

freeze-dried ethanolic leaf extracts and secondary compounds. Foliage from<br />

sugar maple, trembling aspen and mulberry was assayed for possible<br />

insecticidal properties against cells from two lepidopteran defoliators, spruce<br />

budworm and fall armyworm. Cells were suspended in buffered agarose and<br />

spread in a petri dish. Leaf extracts solubilized in 50-70% DMSO were<br />

applied directly to the cells. Trypan blue staining was used as an indicator of<br />

toxicity. Quantitative comparisons were determined by threshold doses that<br />

elicited positive responses. Toxicity and validity of the assay were further<br />

confirmed by the presence of membrane disruption and cellular lysis. The<br />

activity of trembling aspen was markedly enhanced when the pH was raised<br />

from 7 to 10.5, a level similar to that of the larval midgut, but the effect was<br />

largely abolished in the presence of gut juice. Analysis of a crude mulberry<br />

extract treated with neat gut juice suggested that most of the active<br />

material in the lepidopteran leaf diet is either insoluble or precipitated in the<br />

larval midgut, while the activity of any solubilized material is suppressed<br />

through interaction with gut-juice proteins.<br />

Molecular entomology using insect cell lines: A spruce budworm<br />

midgut cell line, FPMI-CF-203 has been shown to respond to the molting<br />

hormone ecdysone, juvenile hormone and other chemicals. Not only was<br />

gene expression stimulated in these treated cells, but they allowed for the<br />

analysis of house keeping genes or molting gene promoters as well as the<br />

study of cell signaling pathways such as protein kinase C and its targets,<br />

steroid hormone and molting gene expression. In addition, GFP tag fused<br />

target proteins were readily visible in single living CF-203 cells. They were<br />

permissive for the production of active foreign gene proteins using<br />

recombinant baculoviruses vector systems. We have observed that while<br />

RNA interference (RNAi) technology for the study of gene function does not<br />

work well in vivo within the whole insect, CF-203 cells are an invaluable in<br />

vitro tool for this function.<br />

Challenges within insect tissue culture discipline: Developing insect<br />

cell lines suitable for a specific application is a very slow and challenging<br />

process. Primary cultures started may or may not develop into cell lines.<br />

Those that do might take months and the resulting cells may not be<br />

suitable for a desired function. Of the 4-6 M insect species, we currently<br />

have 800+ cell lines from 100 species. There are presently no cell lines from<br />

exotic or invasive species. Once established, insect cells, like insects, must be<br />

fed to be kept alive. Suitable culture medium for the growth of tissues of<br />

different insects is currently not available. Large scale production of insect<br />

viruses would require the media to be optimal for cell growth as well as for<br />

replication of the virus or other control agents. Each cell line/virus<br />

combination requires its own unique media composition. Two pressing<br />

concerns requiring immediate attention are cross and misidentification<br />

contamination of existing cell lines and retiring of key researchers. With few<br />

insect cell lines in public collections, private collections are often<br />

inaccessible after the principal investigator leaves. Retiring scientist equals<br />

lost insect cell cultures.<br />

Future requirements to achieve “cultural immortalization”: More<br />

dedicated researchers developing new lines from different species,<br />

routine and aggressive identification, characterization and verification of<br />

cells lines employing new molecular techniques and maximizing the<br />

potential of insect cells as viable commercial ventures are needed.<br />

P72<br />

Down-regulation of CD81 in human cells producing HCV-E1/E2<br />

retroVLPs<br />

Ana F Rodrigues 1 , Miguel R Guerreiro 1 , Rute Castro 1 , Hélio A Tomás 1 ,<br />

Charlotte Dalba 2 , David Klatzmann 3 , Paula M Alves 1 , Manuel J T Carrondo 1,4 ,<br />

Ana S Coroadinha 1*<br />

1 Instituto de Biologia Experimental e Tecnológica/Instituto de Tecnologia<br />

Química e Biológica (IBET/ITQB-UNL), Apartado 12, P-2781-901 Oeiras,<br />

Portugal; 2 EPIXIS SA, 5 rue des Wallons, 75013 Paris, France; 3 AP-HP, Hôpital<br />

Pitié-Salpêtrière, Biotherapy, F-75013 Paris, France; 4 Faculdade de Ciências e<br />

Tecnologia/Universidade Nova de Lisboa (FCT/UNL), P-2825 Monte da<br />

Caparica, Portugal<br />

E-mail: avalente@itqb.unl.pt<br />

BMC Proceedings 2011, 5(Suppl 8):P72


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Background: Retroviral based biopharmaceuticals are used in numerous<br />

therapeutic applications, from gene therapy to vaccination [1-3].<br />

Nonetheless, retroviruses are known to incorporate proteins from the host<br />

cell which may increase the vector immunotoxicity [4]. CD81, a membrane<br />

protein belonging to the tetraspanin superfamily has been recently<br />

identified as one of the host cell proteins majorly incorporated into Moloney<br />

Murine Leukemia Virus (MoMLV) derived vectors[5]. Therefore, CD81 was<br />

chosen as a target for studying the effects of host cell protein incorporation<br />

in the immunotoxic profile of retroviral vectors (RV). A human cell line,<br />

293rVLP, producing retrovirus like particles (rVLPs) was used to prove the<br />

concept of customizing RV composition by manipulating cellular protein<br />

content, using the CD81 tetraspanin as a case study. rVLPs will be used as<br />

candidate vaccines for hepatitis C by displaying HCV-E1/E2 proteins.<br />

Materials and methods: Cell lines and culture media: 293rVLP is a HEK<br />

293 (ATCC CRL-1573) based cell line established by stable transfection of<br />

pCeB plasmid [6] driving MoMLV gag-pol expression as described in [7].<br />

293T cells (ATCC CRL-11268) were used for producing and titer shRNA<br />

lentiviral vector stocks. All cells were maintained in Dulbecco’s modified<br />

Eagle’s medium (DMEM) (Gibco) 10% (v/v) Foetal Bovine Serum (FBS)<br />

(Gibco).<br />

Plasmids and lentivirus production: Short hairpin (sh) RNA lentiviral vectors<br />

based on pLKO.1-puro [8] containing a puromycin resistance gene and<br />

targeting five different sequences on the human CD81 mRNA or a nontarget<br />

(nt) shRNA vector were purchased from Sigma Mission® RNAi (Sigma).<br />

The lentiviral vector stocks were produced by transient co-transfection of<br />

293T cells with pSPAX2, pMD2G (Addgene) and the respective pLKO.1-puroshRNA.<br />

Flow cytometry for CD81 detection: The presence of CD81 on the cell<br />

membrane was detected by flow cytometry using mouse monoclonal<br />

antibody against human CD81 and an Alexa FluorR 488 dye conjugated<br />

secondary antibody (Invitrogen).<br />

Oxidative stress induction and cell viability: The susceptibility to oxidative<br />

stress injury was assessed by analyzing tert-butyl hydroperoxide induced<br />

death. Cell viability was quantified by the percentage of cells staining<br />

negative for PI (by flow cytometry).<br />

RT activity: To determine MoMLV reverse transcriptase (RT) activity the<br />

RetroSys C-Type RT activity kit (Innovagen) was used.<br />

Further details on Materials and Methods are in [9].<br />

Results and discussion: 293rVLP cells producing rVLPs were used to<br />

evaluate the possibility of customizing vector composition by<br />

manipulating host protein content. CD81 tetraspanin was chosen to be<br />

down-regulated by shRNA, given that it is significantly incorporated into<br />

Page 102 of 181<br />

RV particles, conferring them a strong immunogenic character in mice.<br />

Each cell population, shCD81 8 to 12, as well as the non-target control,<br />

were analyzed for the presence of CD81 by flow cytometry (Table 1).<br />

CD81 down-regulation efficiency varied considerably among the five<br />

shRNA sequences, ranging from negligible in the case of shCD81 11 to a<br />

very strong silencing effect in shCD81 10 cell population, with more than<br />

90% of cells being negative for CD81. ntshRNA population stained almost<br />

totally positive (94%) for CD81. To increase CD81 down-regulation, the<br />

shCD81 10 cell population was doubly infected with shCD81 9 and 12<br />

(Table 1). Only 1.5% of the resulting cells were positive for CD81 which,<br />

considering the background values for the ntshRNA control (approx. 6%),<br />

suggested a totally CD81 silenced population.<br />

shCD81 9+10+12 cell population was cloned by limiting dilution and 30<br />

clones were isolated. Four of those, #14, #19, #22 and #30 were chosen as<br />

representatives of the highest CD81 silencing (≥85%) and #9 as a<br />

representative of intermediate CD81 silencing. The rVLP production was<br />

analyzed by reverse transcriptase (RT) quantification in the supernatant; the<br />

cellular response to CD81 down-regulation and/or RNAi pathway activation<br />

was assessed by evaluating cell growth and cell death susceptibility under<br />

oxidative stress conditions (Table I). Cell population ntshRNA was used as<br />

negative control to distinguish CD81 silencing from RNAi pathway activation<br />

effects. The results were compared to the parental cell line, 293rVLP. With<br />

the exception of clone #14, all silenced clones presented lower cell growth.<br />

This effect, seemed to be attributed to CD81 down-regulation, as it was not<br />

detected in the ntshRNA control. Indeed, CD81 has been previously<br />

described to be implicated in proliferation and cytostasis of various cell<br />

types, including lymphocytes, fibroblasts, epithelial cells and astrocytes<br />

[10-13]. Lower half maximal inhibitory concentration values (IC 50) under<br />

oxidative stress were observed for silenced cells, including the ntshRNA<br />

population, suggesting that the activation of RNAi pathway played a role in<br />

injury induced death susceptibility. From a bioprocessing point of view,<br />

increased susceptibility to stressful conditions, oxidative or other, should be<br />

a drawback. However, RNAi is faster and simpler than a knockout, and<br />

undeniably useful in preliminary stages. Additionally, for certain vital<br />

proteins, a total knock-out may not be possible.<br />

To assess rVLP productivity in shCD81 clones the RT in the viral supernatant<br />

was quantified. Three of the five clones presented lower RT productivities,<br />

whereas clones #14 and #22 showed identical values to the non-target<br />

silenced control or the parental cell line. This suggested that CD81 downregulation<br />

does not affect viral particles production. Finally, rVLP shCD81<br />

#14, was used to produce rVLPs, which were concentrated, purified and<br />

analyzed for the presence of CD81 by Western blotting. The rVLPs were<br />

Table 1(abstract P72) CD81 down-regulation in 293rVLP cells<br />

CD81 - cells<br />

(%)<br />

sh populations ntshRNA 5.8<br />

shCD81 8 20.4<br />

shCD81 9 88.4<br />

shCD81 10 92.8<br />

shCD81 11 10.0<br />

shCD81 12 31.0<br />

shCD81 9+10<br />

+12<br />

98.5<br />

CD81 - cells Specific cell growth<br />

(%)<br />

(h -1 IC50 (µM tert-butyl<br />

Specific RT productivity<br />

)<br />

hydroperoxide)<br />

(10 -6 ng/cell.h)<br />

293 rVLP 2.1 0.0201 ± 0.0002 3.3 ± 1.1 1.7 ± 0.3<br />

ntshRNA 0.8 0.020 ± 0.002 9.2 ± 1.4 2.3 ± 0.4<br />

shCD81 9+10+12<br />

clones<br />

shCD81 #9 86.4 0.0124 ± 0.0004 15.4 ± 1.0 0.9 ± 0.2<br />

shCD81 #14 97.1 0.021 ± 0.002 4.5 ± 1.1 2.0 ± 0.4<br />

shCD81 #19 92.9 0.015 ± 0.001 6.5 ± 1.1 1.2 ± 0.2<br />

shCD81 #22 88.9 0.018 ± 0.001 6.0 ± 1.1 1.8 ± 0.4<br />

shCD81 #30 82.3 0.017 ± 0.001 5.6 ±1.0 1.4 ± 0.2


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

shown to be absent of CD81 demonstrating that it could be eliminated.<br />

These results suggest that, although naturally incorporated on RV, CD81 is<br />

notessentialfortheassemblyorsecretion of fully functional particles.<br />

Hepatitis C envelope proteins E1/E2 were successfully expressed in CD81<br />

silenced cells and incorporated in rVLPs.<br />

Conclusions: In this work it was demonstrated that is possible to<br />

manipulate the composition of the host cell proteins incorporated into<br />

rVLPs. rVLps can be used as a epitope display platform and used in vaccine<br />

development therefore, CD81 was chosen since it is strongly immunogenic<br />

in mice. The results herein presented support that is possible to manipulate<br />

cellular protein expression, CD81 or others, as a tool to control RV<br />

composition and their immunogenic profile. Additionally, for gene therapy<br />

purposes, the removal immunotoxic proteins in the producer cell may<br />

improve the in vivo efficacy of retrovirus, lentivirus or other enveloped virus.<br />

Acknowledgements: The authors acknowledge the financial support<br />

received from the European Commission (Clinigene: LSHB-CT2006-018933)<br />

andfromFundaçãoparaaCiênciaeaTecnologia(FCT)—Portugal (PTDC/<br />

EBB-BIO/102649/2008 and PTDC/EBB-BIO/100491/2008). A.F. Rodrigues<br />

acknowledge FCT for her PhD grant (SFRH/BD/48393/2008).<br />

References<br />

1. Dalba C, Bellier B, Kasahara N, Klatzmann D: Replication-competent vectors<br />

and empty virus-like particles: new retroviral vector designs for cancer<br />

gene therapy or vaccines. Mol Ther 2007, 15:457-466.<br />

2. Dalba C, Klatzmann D, Logg CR, Kasahara N: Beyond oncolytic virotherapy:<br />

replication-competent retrovirus vectors for selective and stable<br />

transduction of tumors. Curr Gene Ther 2005, 5:655-667.<br />

3. Edelstein ML, Abedi MR, Wixon J: Gene therapy clinical trials worldwide to<br />

2007–an update. J Gene Med 2007, 9:833-842.<br />

4. Arthur LO, Bess JW Jr., Urban RG, Strominger JL, Morton WR, et al:<br />

Macaques immunized with HLA-DR are protected from challenge with<br />

simian immunodeficiency virus. J Virol 1995, 69:3117-3124.<br />

5. Segura MM, Garnier A, Di Falco MR, Whissell G, Meneses-Acosta A, et al:<br />

Identification of host proteins associated with retroviral vector particles<br />

by proteomic analysis of highly purified vector preparations. J Virol 2008,<br />

82:1107-1117.<br />

6. Danos O, Mulligan RC: Safe and efficient generation of recombinant<br />

retroviruses with amphotropic and ecotropic host ranges. Proc Natl Acad<br />

Sci U S A 1988, 85:6460-6464.<br />

Table 1(abstract P73) Characteristics of the different single-use depth filters<br />

7. Cosset FL, Takeuchi Y, Battini JL, Weiss RA, Collins MK: High-titer packaging<br />

cells producing recombinant retroviruses resistant to human serum.<br />

J Virol 1995, 69:7430-7436.<br />

8. Moffat J, Grueneberg DA, Yang X, Kim SY, Kloepfer AM, et al: A lentiviral<br />

RNAi library for human and mouse genes applied to an arrayed viral<br />

high-content screen. Cell 2006, 124:1283-1298.<br />

9. Rodrigues AF, Guerreiro MR, Santiago VM, Dalba C, Klatzmann D, et al:<br />

Down-regulation of CD81 tetraspanin in human cells producing<br />

retroviral-based particles: tailoring vector composition. Biotechnol Bioeng<br />

2011, 108(11):2623-2633.<br />

10. Geisert EE Jr., Abel HJ, Fan L, Geisert GR: Retinal pigment epithelium of<br />

the rat express CD81, the target of the anti-proliferative antibody<br />

(TAPA). Invest Ophthalmol Vis Sci 2002, 43:274-280.<br />

11. Geisert EE Jr., Yang L, Irwin MH: Astrocyte growth, reactivity, and the target<br />

of the antiproliferative antibody, TAPA. J Neurosci 1996, 16:5478-5487.<br />

12. Oren R, Takahashi S, Doss C, Levy R, Levy S: TAPA-1, the target of an<br />

antiproliferative antibody, defines a new family of transmembrane<br />

proteins. Mol Cell Biol 1990, 10:4007-4015.<br />

13. Toledo MS, Suzuki E, Handa K, Hakomori S: Cell growth regulation through<br />

GM3-enriched microdomain (glycosynapse) in human lung embryonal<br />

fibroblast WI38 and its oncogenic transformant VA13. J Biol Chem 2004,<br />

279:34655-34664.<br />

P73<br />

Evaluation of disposable filtration systems for harvesting high cell<br />

density fed batch processes<br />

Antje Pegel * , Friedemann Übele, Sven Reiser, Dethardt Müller, Gregor Dudziak<br />

Rentschler Biotechnologie GmbH, 88471 Laupheim, Germany<br />

E-mail: antje.pegel@rentschler.de<br />

BMC Proceedings 2011, 5(Suppl 8):P73<br />

Introduction: In the underlying study we evaluated different single-use<br />

filtration systems for cell separation and harvest clarification in 1,000 L<br />

scale. A screening of different depth filters was carried out with various<br />

single-use filters from Pall, Cuno (3M), Millipore and Sartorius Stedim. In<br />

total, we included 85 depth filtrations in the screening. Out of that, two<br />

single-use filtration systems were chosen and further tested in 200 L<br />

Manufacturer Material* Filter type Retention range (µm)* Number of filter layers*<br />

PDK7 20 – 4<br />

Pall<br />

Seitz® HP-Series<br />

Cellulose,<br />

Diatomaceous earth,<br />

Resin<br />

PDK6 20 – 3<br />

PDK5 20 – 1 2<br />

PDH4 15 – 0.4<br />

PDE2 3.5 – 0.2<br />

Seitz® P-Series KS50P 0.8 – 0.4 1<br />

10SP02A 7 – 1<br />

Cuno<br />

Zeta Plus®<br />

Millipore<br />

Millistak+®<br />

Sartorius<br />

Sartoclear P®<br />

Cellulose,<br />

Diatomaceous earth,<br />

Perlite<br />

Cellulose,<br />

Diatomaceous earth<br />

Cellulose,<br />

Diatomaceous earth,<br />

Binding matrix<br />

30SP02A 5 – 0.8 2<br />

60SP02A 5 – 0.65<br />

60ZA05A 0.8 – 0.6<br />

D0HC 9 – 0.6 2<br />

C0HC 2 – 0.2<br />

PB1 11 – 4 2<br />

PB2 8 – 1<br />

* Data according to manufacturers [http://www.pall.com, http://www.cuno.com, http://www.millipore.com, http://www.sartorius-stedim.com].<br />

Page 103 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

scale. Based on these results, a single-use filtration set-up for harvesting<br />

production scale fed batch processes was determined.<br />

Material and methods: High cell density fed batch cultivations of a<br />

monoclonal antibody (mAb) expressing Chinese Hamster Ovary (CHO) cell<br />

line were harvested by depth filtration and 0.2 µm filtration after 14 to<br />

Page 104 of 181<br />

19 days at viabilities ranging from 40 to 95%. For the screening in 10 L<br />

scale, single-use depth filters (23 to 26 cm 2 ) with different separation<br />

ranges were used (Table 1). Subsequently, two disposable depth filtration<br />

systems were tested in 200 L scale using filter capsules with a filter area of<br />

0.23 to 0.25 m 2 . Depth filtrates were 0.2 µm filtered with Pall EKV (20 cm 2 ).<br />

Figure 1(abstract P73) A) Maximum capacities of depth filters in small scale screening. B) Calculated filter areas for filtration of 1,000 L harvest based on<br />

results of large scale trials.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

During filtration, a constant flow of 100 L·m -2 ·h -1 was applied. Maximum<br />

capacities of the filters were determined at a pressure of 1 bar. Filter<br />

performance was assessed with regard to filter capacity, filtrate turbidity<br />

and product yield. Furthermore, content of DNA and Host Cell Protein<br />

(HCP) in filtrates were measured.<br />

Results:<br />

Depth filter screening: The filters PDK7, PDK6 and PDK5 from Pall<br />

showed the highest maximum capacities with 161-167 L/m 2 (Figure 1 A).<br />

These double layered filters had identical first membranes and differing<br />

finer second membranes. Filtrate turbidity was below 7 NTU when<br />

applying PDK6, whereas in filtrates generated with the coarser filter PDK7<br />

turbidities up to 9 NTU were observed. Additionally, product loss with<br />

PDK6 (6%) was lower compared with PDK7 (8%) or PDK5 (13%). For that<br />

reason Pall PDK6 was selected for the scale-up experiments.<br />

Additionally, the depth filters 10SP02A and 60SP02A from Cuno were chosen.<br />

For filter 10SP02A a maximum capacity of 131 L/m 2 was obtained. However,<br />

the filtrate turbidity was higher than 10 NTU causing a fast blocking of the<br />

0.2 µm filter. Therefore, this filter was combined with the finer depth filter<br />

60SP02A. The filter 60SP02A was selected due to turbidity values below 10<br />

NTU and an acceptable capacity of 105 L/m 2 when applied stand-alone.<br />

Turbidity breakthroughs at pressures below 1 bar were observed for the<br />

depth filters Millipore D0CH and Sartorius Stedim PB1 (Figure 1 A).<br />

Consequently, these filters were not considered for the scale-up studies.<br />

Scale-up: The selected depth filters were applied in 200 L scale using the<br />

Stax Disposable Depth Filter System (Pall) and the Zeta Plus Encapsulated<br />

System (Cuno), respectively. Performance of depth filters was comparable in<br />

large scale and small scale. Maximum capacities in the large scale trials were<br />

174 L/m 2 for Pall PDK6, 152 L/m 2 for Cuno 10SP02A, and 137 L/m 2 for Cuno<br />

60SP02A. Product loss was below 10%.<br />

Based on maximum capacities filter areas were calculated for harvest in<br />

1,000 L scale (Figure 1 B). An optimal filtration set-up leading to the lowest<br />

total filter area (6.9 m 2 ) was found in the depth filter Pall PDK6 and a<br />

subsequent 0.2 µm filter. Insertion of a second depth filter after Pall PDK6<br />

reduced the area of the 0.2 µm filter but did not affect the total filter area.<br />

The depth filter area of Cuno 60SP02A (7.3 m 2 ) was comparable to that of<br />

the filter combination Cuno 10SP02A – 60SP02A (7.4 m 2 ).<br />

DNA content in the filtrate was reduced by 30% with Pall PDK6 and even<br />

by 70% with the additional depth filter PDE2. The Cuno depth filter<br />

combination 10SP02A – 60SP02A reduced DNA content by 99%<br />

compared to 90% when only applying 60SP02A. With the Pall depth<br />

filters a reduction of HCP by 30% was measured whereas no HCP<br />

removal was observed for the Cuno depth filters.<br />

Conclusion: In this concept study disposable filtration systems were<br />

successfully tested in terms of identifying suitable filtration set-ups for<br />

equipping a 1,000 L disposable manufacturing line. These single-use<br />

filtration systems can thereby replace conventional (stainless steel) disc<br />

centrifuge and filtration steps in industrial mammalian cell culture<br />

production processes. The Stax system from Pall equipped with the filter<br />

PDK6 followed by a 0.2 µm filtration was identified as the first choice singleuse<br />

filtration set-up offering high capacities and a low product loss. Addition<br />

of a finer second depth filter can further increase filtrate clarification<br />

resulting in a reduction of DNA and HCP in filtrates and a smaller area of the<br />

subsequent 0.2 µm filter, but is also combined with a higher risk of product<br />

loss and an increased time and handling effort.<br />

Acknowledgement: We thank Novartis Pharma AG for supporting this<br />

project.<br />

P74<br />

Osteogenic Differentiation of adipose mesenchymal stem cells with<br />

BMP-2 embedded microspheres in a rotating bed bioreactor<br />

Stefanie Boehm 1* , Yael Lupu 2 , Marcelle Machluf 2 , Cornelia Kasper 1<br />

1 Institute of Technical Chemistry, Leibniz University of Hannover, Callinstr. 5,<br />

30167 Hannover, Germany; 2 Faculty of Biotechnology and Food Engineering,<br />

Technion, Haifa, Israel<br />

BMC Proceedings 2011, 5(Suppl 8):P74<br />

Introduction: Bone tissue engineering aims at the generation of functional<br />

bone tissue for replacement of defect bone tissue in order to reestablish<br />

normal function. For this purpose mesenchymal stem cells (MSCs) are widely<br />

used since they can be isolated from different sources and can easily be<br />

differentiated in vitro into the mesenchymal lineages [1,2].<br />

Page 105 of 181<br />

The aim of this work was to study the effect of growth factor BMP-2 on<br />

the osteogenic differentiation of human mesenchymal stem cells from<br />

adipose tissue. The cells were cultivated in a rotating bed bioreactor<br />

system (Z®RPD system, Zellwerk GmbH) on the aluminium oxide based<br />

ceramic Sponceram® for four weeks. The ceramic discs were loaded with<br />

PLGA microspheres releasing BMP-2. For this experiments a system was<br />

used which allows the parallel cultivation of four bioreactors. The used<br />

bioreactor containers were manufactured of disposable material for single<br />

use application. During the cultivation glucose and lactate concentrations<br />

were measured and after the experiments histological stainings were<br />

performed (DAPI, von Kossa and alizarine red). Furthermore the<br />

concentration of alkaline phosphatase was measured in medium samples<br />

and mRNA of the cells was isolated to perform RT-PCR to investigate the<br />

expression of different bone markers.<br />

Materials and methods: Biomaterial: The Sponceram® biomaterial<br />

consists of AlO 2. It has a macroporous structure (600 µm) with a<br />

microporous surface and a porosity of 85%. For the cultivations scaffolds<br />

with a diameter of 65 mm and a thickness of 3 mm were used. The PLGA<br />

microspheres were produced by solvent evaporation and at a size of about<br />

50 µm. The BMP-2 concentration in the microspheres was about 0.05 µg<br />

per mg PLGA. The total amount of BMP-2 in the dynamic cultivations was<br />

2.5 µg in each bioreactor. The BMP-2 release and the concentration in the<br />

medium was measured with a BMP-2 ELISA (Quantikine, R&D Systems) The<br />

BMP-2 concentration in medium samples of bioreactor 4 decreases almost<br />

linear from 875 pg/ml to zero pg/ml in the first 16 days of the dynamic<br />

cultivation.<br />

Cells: The adipose tissue derived mesenchymal stem cells (adMSC) were<br />

isolated by enzymatic treatment under GMP conform conditions in a<br />

laboratory of the Red Cross in Linz, Austria. For the dynamic cultivation the<br />

isolated cells were expanded in cell factories (Nunclon Δ Cell Factory) and<br />

the dynamic cultivations were performed with cells in passage 5. For the<br />

cultivations 5x10 6 cells were used in every bioreactor.<br />

Bioreactor: The Z®RPD bioreactor system consists of four disposable<br />

rotating bed bioreactors and is working in a perfusion mode. The reactor is<br />

equipped with a Sponceram® ceramic, which is served as a rotating bed. In<br />

the standard operation mode the bioreactor is half filled with media (50 mL)<br />

and the Sponceram® is rotating with 0.5 rpm. This way the cells on the<br />

ceramics have alternating contact to medium and gas-filled headspace<br />

improving cell nutrition and oxygen supply.Theaimoftheparallel<br />

cultivation of four bioreactors was, to test the reproducibility of the system,<br />

which is important for GMP standards in clinical application. The bioreactors<br />

were integrated in a GMP conform breeder with a control unit for GMP<br />

conform documentation and evaluation of pH, oxygen and temperature.<br />

Furthermore a sterile sampling of media during the cultivation is possible.<br />

The dynamic cultivations were performed over a time period of 28 days and<br />

the cells were cultivated in osteogenic medium with dexamethasone,<br />

l-ascorbate and b-glycerol phosphate.<br />

Results: During 28 days of dynamic cultivation about 200 mg glucose were<br />

consumed in each bioreactor. The glucose consumption increased during<br />

the cultivation indicating a continuous cell growth on the ceramics. Also<br />

DAPI staining of the cell seeded ceramics showed a high density of cells on<br />

the scaffolds. Furthermore the concentration of alkaline phosphatase was<br />

measured daily (Sigma fast TM , Sigma Aldrich). The amount of the secreted<br />

alkaline phosphatase decreased during the cultivation period which is an<br />

indication for advanced osteogenic differentiation. Also the expression of<br />

various genes involved in the osteogenic differentiation of cells was verified.<br />

Different intensities of the gene expression in the four bioreactors was<br />

observed; especially the gene expression in bioreactor 1 was obviously<br />

higher. SEM pictures of the cell seeded ceramics showed thick cell layers on<br />

Sponceram®. Small round beads and a fibrous structure were noticeable.<br />

The beads might be cells, which are embedded in the extracellular matrix<br />

and the fibrous structure could be a hint for Collagen reposition. To verify<br />

the osteogenic differentiation of the cells von Kossa- and alizarine red<br />

stainings were performed. Both stainings showed high amounts of calcified<br />

matrix on all four ceramics. Also the alkaline phosphatase staining (Sigma<br />

fast BCIP, Sigma Aldrich) was successful and showed high amounts of<br />

secreted alkaline phosphatase on all four scaffolds.<br />

Conclusion: The glucose consumption increased during the cultivation<br />

indicating a continuous cell growth and the consumption was almost<br />

the same in all bioreactors. Also the DAPI staining of the ceramics<br />

showed a homogenous spreading of the cells on the Sponceram®<br />

ceramics.But only in two bioreactors a typical alkaline phosphatase


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P74) ZRP® bioreactor system (Zellwerk GmbH) with the special setup for performing four parallel cultivations.<br />

concentration curve could be verified in collected medium samples. The<br />

histological staining showed matrix calcification of the cells on every<br />

ceramic. Also the expression of typical bone markers was shown. In<br />

summary, osteogenic differentiation of adMSC on Sponceram®, caused<br />

by BMP-2 released from PLGA microspheres, was demonstrated and the<br />

reproducibility of the osteogenic differentiation in four parallel<br />

cultivations was demonstrated.<br />

Acknowledgements: This project is sponsored by the state of lower<br />

Saxony. Moreover we gratefully thank Zellwerk GmbH for providing the<br />

Z®RPD system and Sponceram® ceramics.<br />

References<br />

1. Moretti P, Hatlapatka T, Marten D, Lavrentieva A, Majore I, Hass R, Kasper C:<br />

Mesenchymal stromal cells derived from human umbilical cord tissues:<br />

primitive cells with potential for clinical and tissue engineering<br />

applications. Adv Biochem Eng Biotechnol 2010, 123:29-54.<br />

2. Hass R, Kasper C, Bohm S, Jacobs R: Different populations and sources of<br />

human mesenchymal stem cells (MSC): A comparison of adult and<br />

neonatal tissue-derived MSC. Cell Commun Signal 2011, 9(1):12.<br />

P75<br />

Medium and feed optimization for fed-batch production of a<br />

monoclonal antibody in CHO cells<br />

Nadine Kochanowski * , Gaetan Siriez, Sarah Roosens, Laetitia Malphettes<br />

Cell Culture Process Development Group, Biological Process Development,<br />

UCB Pharma S.A., Braine L’Alleud, 1420, Belgium<br />

E-mail: Nadine.Kochanowski@ucb.com<br />

BMC Proceedings 2011, 5(Suppl 8):P75<br />

Background: Mammalian cells are used extensively in the production<br />

of recombinant proteins, and of monoclonal antibodies (MAbs) in<br />

particular. The trend towards avoiding animal-derived components in<br />

biopharmaceutical production processes has led to the extensive use of<br />

non-animal origin hydrolysates such as plant hydrolysates or yeast<br />

Page 106 of 181<br />

hydrolysates. The source of hydrolysates affects cell growth and productivity<br />

and may also affect product quality. Accordingly, careful consideration<br />

should be given during process and cell culture media development, in<br />

order to determine the appropriate type and amount of hydrolysates to be<br />

added, for the cell and product at hand.<br />

In this study, we assessed the impact of several hydrolysate additives and<br />

chemically defined (CD) commercial feeds on MAb titers, MAb average<br />

specific productivity (average Qp), cell viabilities and metabolite profiles<br />

in suspension cultures of recombinant CHO cells expressing a monoclonal<br />

antibody in shake flasks and 2 L bioreactors.<br />

Materials and methods: Initial experiments were performed using<br />

chemically defined culture medium in 125 mL shake flasks with 50 mL<br />

working volume. CHO cells were seeded at 0.3x10 6 viable cells/mL and<br />

incubated at 140 rpm, 36.5°C and 5% CO2. 2L stirred tank bioreactors<br />

(Sartorius) were carried out for 14 days in a fed-batch mode in a chemically<br />

defined medium supplemented with chemically defined feeds and<br />

hydrolysates. Glucose was maintained between 1 and 6 g/L. At the day of<br />

harvest the clarification was performed by depth filtration. Analysis of daily<br />

samples included determinations of cell viability, cell density, metabolites,<br />

osmolality and product titer. Product concentration of the supernatant<br />

samples was quantified using Octet QK and Protein A high performance<br />

liquid chromatography (HPLC). Protein characterization of Protein-A purified<br />

samples were profiled by reduced and non reduced SDS PAGE. Isoelectric<br />

focusing (IEF) analysis of Protein-A purified MAb was carried out using a<br />

iCE280 IEF Analyzer. Aggregates and monomers proportion were<br />

determined by using size exclusion chromatography. Acidic and basic<br />

species were characterized using anion exchange (AEX) HPLC. Oligosaccharides<br />

were cleaved enzymatically using N-Glycanase, then labeled<br />

with 2-aminobenzamide and analyzed by HPLC using an amide column and<br />

a fluorescent detector.<br />

Results: Several chemically defined feeds and hydrolysates were assessed<br />

on CHO cells expressing a monoclonal antibody in fed-batch mode. The<br />

performance of the developed process was compared to an existing inhouse<br />

platform process.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P75) Chemically defined feeds and hydrolysates tested<br />

Supplier Commercial feed name Feed name in the poster<br />

ThermoFischer Cell Boost 1 CD Feed 1<br />

ThermoFisher Cell Boost 2 CD Feed 2<br />

ThermoFisher Cell Boost 3 CD feed 3<br />

ThermoFischer Cell Boost 4 CD Feed 4<br />

ThermoFischer Cell Boost 5 CD Feed 5<br />

ThermoFischer Cell Boost 6 CD Feed 6<br />

Life Tech CHO Feed A CD Feed 7<br />

Life Tech CHO Feed B CD Feed 8<br />

Life Tech CHO Feed C CD Feed 9<br />

BD Biosciences Yeast Extract Hydrolysate 1<br />

BD Biosciences Yeastolate Hydrolysate 2<br />

BD Biosciences Select Phytone Hydrolysate 3<br />

BD Biosciences Ultrapep Soy Hydrolysate 4<br />

Sheffield HyPep 1510 Hydrolysate 5<br />

Sheffield HyPep 4605 Hydrolysate 6<br />

BD Biosciences 3 g/L Yeast Extract + 3.25 g/L Yeastolate Hydrolysate combination 1<br />

BD Biosciences 3.5 g/L Yeast Extract + 2.75 g/L Yeastolate Hydrolysate combination 2<br />

BD Biosciences 4 g/L Yeast Extract + 2.25 g/L Yeastolate Hydrolysate combination 3<br />

BD Biosciences 4.5 g/L Yeast Extract + 1.75 g/L Yeastolate Hydrolysate combination 4<br />

BD Biosciences 5 g/L Yeast Extract + 1.25 g/L Yeastolate Hydrolysate combination 5<br />

BD Biosciences 3 g/L Yeast Extract + 5.1 g/L Yeastolate Hydrolysate combination 6<br />

BD Biosciences 3.5 g/L Yeast Extract + 4.6 g/L Yeastolate Hydrolysate combination 7<br />

BD Biosciences 4 g/L Yeast Extract + 4.1 g/L Yeastolate Hydrolysate combination 8<br />

BD Biosciences 4.5 g/L Yeast Extract + 3.6 g/L Yeastolate Hydrolysate combination 9<br />

BD Biosciences 5 g/L Yeast Extract + 3.1 g/L Yeastolate Hydrolysate combination 10<br />

Page 107 of 181<br />

Figure 1(abstract P75) Relative percentage of improvement on MAb titer. Note: Platform process in shake flask was used as the 100% reference for all<br />

the calculations of relative percentage of improvement on Mab titer measured the day of harvest.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Nine different chemically defined feeds were assessed and added at<br />

different concentrations (Table 1). Among the feeds tested, addition of<br />

CD Feed 8 and 9 brought a 150% improvement on MAb titer on the day<br />

of harvest compared to the platform process. All the MAb titers<br />

measured were ranging from 2 g/L to 6 g/L (Figure 1).<br />

Six different hydrolysates were assessed at different concentrations in fedbatch<br />

mode (Table 1). Among the feeds tested, addition of hydrolysate 1<br />

and hydrolysate 2 showed an improvement of 175% and 167%<br />

respectively on MAb yield.<br />

To identify potential synergies between hydrolysates, the best<br />

hydrolysates from previous experiments were selected and were tested in<br />

combination at different ratios on CHO cell cultures (Table 1). Antibody<br />

concentration at harvest was 290% higher with some of the hydrolysate<br />

combinations.<br />

Based on feed combination optimization results, the number of bolus<br />

feeds and the feed addition timing were then fine-tuned using the best<br />

hydrolysate combination. Reducing the number of bolus feeds enabled to<br />

reduce ammonia and osmolality while maintaining a high MAb titer<br />

(Figure 1). Moreover, under these conditions, cell viabilities were<br />

maintained above 80% throughout the culture (data not shown).<br />

Based on experimental results obtained in shake flasks, the best hydrolysate<br />

combination (3 feeds and 4 feeds) and CD feed were assessed on CHO cells<br />

cultured in 2 L stirred tank bioreactors. Cell growth and cell metabolism<br />

were monitored daily throughout the cultures in bioreactors. By feeding the<br />

cultures with hydrolysates, addition of 3 or 4 bolus feeds enabled to attain<br />

similar maximum viable cell count. Addition of chemically defined feed led<br />

to a 30% higher maximum viable cell count. Cell viabilities were maintained<br />

at acceptable values throughout the cultures in the established culture<br />

conditions. Lactate profiles were similar independently of the feeding<br />

regime. Decreasing the number of hydrolysate feeds enabled to maintain<br />

osmolality and ammonia at acceptable concentrations for CHO cell growth<br />

and product quality. Cell growth performance and metabolism profiles<br />

observed in 2 L bioreactors were comparable to those observed in shake<br />

flasks.<br />

Product titers have been measured throughout the fed-batch cultures with<br />

the Octet QK system. The MAb titers were in a 2-6 g/L range for all the<br />

tested feeding regimes at the day of harvest. A combination of<br />

hydrolysates and a chemically defined feed supplementation showed an<br />

improvement of 296% and 245% on MAb concentration at the day of<br />

harvest in comparison to the platform process (Figure 1). MAb average<br />

specific productivity (Qp) was increased by 700% and 360% by adding 4<br />

feeds of hydrolysates and chemically defined feed respectively. Decreasing<br />

the number of hydrolysate feeds showed a slight decrease on MAb titer<br />

and on Qp.<br />

Product quality attributes were determined on cell culture clarified fluids after<br />

Protein-A purification. Reduced and non reduced SDS electrophoresis,<br />

isoelectric focusing (IEF) analysis, gel permeation HPLC, size exclusion (SEC)<br />

chromatography, anion exchange HPLC (AEX) have been used to characterize<br />

the Protein-A purified MAb. Product quality data was comparable for all the<br />

feeding regimes tested in 2 L bioreactors.<br />

Conclusions: Hydrolysate combination additions significantly improved<br />

MAb production in comparison to single hydrolysate addition or<br />

chemically defined feeds. Number of bolus feeds and feeding timing<br />

optimization enabled us to improve the process robustness taking into<br />

account the impact of feeding strategy on cell metabolism and product<br />

quality. The feeding regimes established in shake flasks led to similar<br />

culture performance in 2 L bioreactors.<br />

Acknowledgments: We wish to acknowledge BD Biosciences, Sheffield,<br />

Life Tech and Thermofisher for providing media and feeds. We also wish<br />

to acknowledge Mari Spitali, Natasha Hinkel, Vanessa Auquier and Marc<br />

Speleers for Protein-A purification and product quality analysis.<br />

P76<br />

In-situ microscopy and 2D fluorescence spectroscopy as online methods<br />

for monitoring CHO cells during cultivation<br />

Sophia Bonk * , Marko Sandor * , Ferdinand Rüdinger, Bernd Tscheschke,<br />

Andreas Prediger, Alexander Babitzky, Dørte Solle, Sascha Beutel,<br />

Thomas Scheper<br />

Institute of Technical Chemistry, Leibniz University of Hanover, Germany<br />

BMC Proceedings 2011, 5(Suppl 8):P76<br />

Page 108 of 181<br />

Introduction: Typical methods for monitoring cultivation processes are<br />

offline analyses like cell counting, measurement of various substrates and<br />

products (e. g. glucose or lactate) as well as the online monitoring of<br />

several physical process parameters (temperature, pH-value or the<br />

concentration of dissolved oxygen).<br />

To improve cell cultivations detailed information about important analytes<br />

should be available online. Therefore new monitoring methods need to be<br />

established, preferably as in-situ methods to minimize the risk of<br />

contamination.<br />

Two different in-situ online-methods were used to monitor cultivations: Insitu<br />

microscopy and 2D fluorescence spectroscopy. Therefore CHO-K1 cells<br />

(provided by University of Bielefeld) were cultivated in a complex culture<br />

medium (TC 42, TeutoCell, Bielefeld, Germany) using a 2.5 L stainless steel<br />

reactor with a work volume of 2 L. A total of three cultivation runs were<br />

conducted.<br />

In-situ microscopy: The in-situ microscope (ISM), developed at our<br />

institute, offers the possibility to determine the cell density and to obtain<br />

further information about certain cell characteristics such as size,<br />

compactness and excentricity. In addition, it is possible to extract other<br />

information like cell clusters or microbial contaminations from the recorded<br />

images. In this way a general comprehensive overview of a cultivation can<br />

be obtained.<br />

The in-situ microscope was immersed directly in the culture liquid using a<br />

25 mm standard connection. During the cultivation process several images<br />

were obtained and subsequently evaluated by a self-developed algorithm<br />

which detects cells on the basis of grey scale values.<br />

It was necessary to correlate the cells per picture with offline data<br />

determined by a Neubauer counting chamber. This correlation gives a<br />

regression coefficient (R 2 )of0.988.<br />

By means of this correlation the cells per picture were calibrated in reference<br />

to the counted cells per mL (root mean square error of calibration; RMSEC:<br />

0.989). As a result it is possible to detect the cell density via ISM with an<br />

average standard error of 0.027 (pictures per cycle: 300) during further<br />

cultivations.<br />

2D fluorescence spectroscopy: The 2D fluorescence spectra were<br />

collected using a Bioview® sensor (Delta, Denmark). The fluorescence<br />

spectroscopy was used to monitor glucose, lactate und glutamate during<br />

cultivation. At first it was necessary to build a calibration model for each<br />

compound. Therefore two process runs were observed by collecting 2D<br />

fluorescence spectra every 15 min. Approximately every 6 hours a sample<br />

was taken and analyzed offline for glucose, lactate and glutamate. The<br />

offline reference data and the corresponding spectra were used to build a<br />

chemometric model for prediction of the most interesting variables during a<br />

further cultivation run.<br />

Utilizing multivariate analyzing software (Unscrambler X, vers. 10.1) a<br />

calibration model was built by partial least square regression (PLS1) for<br />

every variable. The number of factors necessary to built a model as well as<br />

the results for the regression coefficient (R 2 ), the root mean square error of<br />

calibration and validation (RMSEC and RMSEV) for every variable are shown<br />

in Tab. 1.<br />

The PLS-models were used to predict all three compounds in a third<br />

cultivation run. The standard errors of prediction (RMSEP) are also shown<br />

in Tab. 1. For glutamate the RMSEP is about 5% compared to a maximum<br />

glutamate concentration of 0.27 g/L.<br />

By applying the PLS-models to the fluorescence data the concentration<br />

gradients for every single compound can be predicted (Fig. 1).<br />

Conclusions: In biotechnology, especially in pharmaceutical<br />

biotechnology it is important to closely monitor a cultivation process. The<br />

in-situ microscopy together with digital image processing and the 2D<br />

fluorescence spectroscopy in combination with chemometric tools<br />

complement one another as online monitoring methods.<br />

It was demonstrated that the ISM can be used to monitor the cell density<br />

during a bioprocess with the same accuracy in comparison to a Neubauer<br />

counting chamber. After a successful calibration the cell density can be<br />

determined without the need for taking samples. This can reduce the risk<br />

of contamination, particularly when the cells are cultivated without the<br />

addition of antibiotics.<br />

The Bioview® sensor could be utilized to observe three important analytes<br />

during a cultivation run with an error of prediction under 10%. By<br />

monitoring these analytes in real time it is possible to observe sudden or<br />

unexpected changes during a cultivation and if necessary to react<br />

accordingly.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P76) PLS-model results for glucose, lactate and glutamate<br />

Compound factors R 2<br />

RMSEC [g/L] RMSEV [g/L] RMSEP [g/L]<br />

Glucose 3 0.967 0.381 0.468 0.524<br />

Lactate 3 0.972 0.368 0.461 0.494<br />

Glutamate 4 0.983 0.0036 0.0059 0.0155<br />

Figure 1(abstract P76) Predicted concentration gradients and their corresponding offline values.<br />

Both online methods were successfully applied to monitor the cell density<br />

as well as the glucose, lactate and glutamate concentration during<br />

cultivation.<br />

Outlook: Concerning the ISM further research is focused on a new<br />

prototype with precise linear stages makingitpossibletoadjusttoa<br />

certain sample volume. This should make a calibration with offline data<br />

unnecessary.<br />

The robustness of the calibration models for the 2D fluorescence<br />

spectroscopy will be tested by adding glucose during cultivation in order<br />

to determine if the model can be applied to a fed batch cultivation.<br />

Acknowledgement: This project was funded by the Federal Ministry of<br />

Education and Research within the project “SpektroSens” (spectroscopic<br />

sensor systems for the area of biotechnology and food).<br />

P77<br />

On-line and real time cell counting and viability determination for<br />

animal cell process monitoring by in situ microscopy<br />

Philipp Wiedemann 1,2* , Markus Worf 2 , Hans B Wiegemann 2 , Florian Egner 4 ,<br />

Christian Schwiebert 4 , Jeff Wilkesman 5 , Jean S Guez 3 , Juan C Quintana 2 ,<br />

Diego Assanza 2 , Hajo Suhr 2<br />

1 At present: School of Biotechnology and Biomolecular Sciences, University<br />

of New South Wales, Sydney NSW 2052, Australia; 2 Mannheim University of<br />

Applied Sciences, Paul-Wittsack-Str.10, D-68163 Mannheim, Germany;<br />

3 Laboratoire ProBioGEM, UPRES-EA 1024, Polytech-Lille / IUT A, Université<br />

des Sciences et Technologies de Lille, Avenue Paul Langevin, Villeneuve<br />

d’Ascq, F-59655, France; 4 InVivo BioTech Services, Neuendorfstr. 24a, D-16761<br />

Hennigsdorf, Germany; 5 University of Carabobo, Faculty of Sciences and<br />

Technology, Chemistry Department, Valencia, 2005, Venezuela<br />

E-mail: p.wiedemann@hs-mannheim.de<br />

BMC Proceedings 2011, 5(Suppl 8):P77<br />

Page 109 of 181<br />

Background: Two of the key parameters to be monitored during cell<br />

cultivation processes are cell concentration and viability. Until today, this is<br />

very often done off-line by sterile sampling and subsequent counting using<br />

a hemocytometer or an electronic cell counter. Cell biology lacks a<br />

measurable quantity by which single cells in suspension can be noninvasively<br />

diagnosed as dead or alive. However, it would be of significance<br />

for process monitoring and in the light of initiatives like PAT if cell density as<br />

well as viability could be determined directly and on-line.<br />

Optical measurement of cell density by in situ microscopy eliminates the<br />

need for sampling and allows for continuous monitoring of this key<br />

parameter; see e.g. [1,2]; Guez et al. [1] describe an in situ microscope (ISM)<br />

which does not use any moving mechanical parts within or outside the<br />

fermentation vessel. It transmits in real time images taken directly in the<br />

stirred suspension within the bioreactor. Image data is processed and<br />

evaluated to provide monitoring of cell-density and morphological<br />

parameters, e.g. cell size, by means of assessing the obtained in situ cellmicrographs.<br />

Previously, we have extended in situ microscopy towards viability<br />

assessment of suspended cells [3,5]. Now, we present new findings on<br />

this topic and show that in cultures of suspended cells, cell-death<br />

corresponds to measurable changes in morphometric parameters as e.g.<br />

variance, contrast or entropy of the greyvalues of in situ cell-micrographs.<br />

As an example, here we show viability determination via greyvalue<br />

dispersion.<br />

Material and methods: We use a custom built high resolution ISM (HS<br />

Mannheim) with water immersion objective, 40x magnification, numerical<br />

aperture 0.75 equipped with optical fiber illumination. Data acquisition is<br />

at 0.3 – 15 frames per second, frames have 1293x1040 pixels; primary<br />

data analysis results in cell micrographs (Figure 1a). We have applied the<br />

system to bench top and larger bioreactors (see e.g. [4]) and worked<br />

mainly with Jurkat, CHO and Hybridoma cells.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P77) a: In situ micrographs of hybridoma cells. Left: Typical portrait taken in a cell culture with high viability (>90%) Right: Typical portrait<br />

taken in a cell culture with low viability (


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Ethanol at 42 hours. Cell counts (not shown) and viability were<br />

determined by in situ microscopy and, as reference, by means of a ViCell<br />

cell viability analyser (Beckman Coulter) and Flow Cytometry (Partec)<br />

using Annexin V / FITC and PI staining. Jurkat cells (DSMZ ACC 282) were<br />

cultured in 90% RPMI 1640 + 10% FBS.<br />

Results: Figure 1a shows ISM-micrographs and corresponding greyvalue<br />

histograms during a hybridoma culture. The cell on the left side represents<br />

the typical image of cells during exponential growth at high viability. The<br />

cell on the right side represents the typical image of cells during the final<br />

stage of a culture at low viability. Cells at low viability are much less<br />

homogeneous and have a correspondingly wider dispersion of greyvalues<br />

as compared to cells at high viability. Therefore, the greyvalue-variance<br />

constitutes a random variable which is sensitive to the viability of the<br />

culture, meaning that the greyvalue-variances increase when viability drops.<br />

In order to define - on the basis of this effect - a measurable value<br />

analogous to viability, we compute the percentage of cells with greyvaluevariance<br />

below a certain threshold. The threshold is optimized in calibration<br />

experiments applying reference methods for viability determination. We call<br />

this value “percentage of cells at low variance” (PLV). The PLV value is a<br />

viability value with respect to inhomogeneity of in situ cell-micrographs.<br />

This is analogous to viability defined as percentage of living cells with<br />

respect to a specific ex situ diagnosis. It has to be conceded that there is no<br />

one to one correspondence between vital cells and cells with variance<br />

below the defined threshold. Both sets – vital cells on the one side and cells<br />

with low variance on the other side– are not perfectly identical since some<br />

cells may be dead with no strong inhomogeneity, and vice versa. Despite<br />

these statistical side-effects, the PLV signal constitutes an operative measure<br />

of “inhomogeneity” and as such it also reflects the viability of the culture<br />

(see Figure 1a/b). Therefore, a high correlation can be anticipated between<br />

the PLV-signal and the viability signals from e.g. counting via Trypan blue<br />

exclusion or Flow Cytometry with AV/PI assay.<br />

We perform online computation of PLV-values over the course of the<br />

culture to continuously monitor ISM-viability of the culture in addition to<br />

cell density (see [1]; [4]). Since data analysis and image processing<br />

happens continuously online, micrograph portraits of cells can also be<br />

assessed for other morphological characteristics as for example cell-size in<br />

real time.<br />

Figure 1c shows results of this viability determination in the case of a<br />

Jurkat culture as an example. Similar results have been obtained with CHO<br />

and hybridoma cells. The culture was insulted by addition of 3% Ethanol to<br />

assess responsiveness of the in situ microscope. Similar correlations of<br />

viability determination by in situ microscopy and reference methods were<br />

obtained after addition of Etoposide (final conc. 10µM), infliction of<br />

osmotic shock or no culture insult at all. A very good correlation between<br />

viability determination by ISM, ViCell (i.e. Trypan Blue) and Flow Cytometry<br />

is observed (Figure 1c).<br />

Conclusions and perspectives: We demonstrate that our ISM is suitable<br />

for determination of suspension cell density AND viability on-line and in<br />

real time. Future work will include investigating how the fine-structure in<br />

micrographs and variance or entropy histograms corresponds to specific<br />

apoptotic stages.<br />

For research and process applications, this work introduces non-invasive<br />

live/dead-classification of suspended mammalian cells in real time.<br />

Acknowledgements: P.W. and H.S. are funded by the Karl Völker<br />

Stiftung, Mannheim, and Land Baden-Württemberg. We gratefully<br />

acknowledge the support of Ariane Tomsche, HS Mannheim.<br />

References<br />

1. Guez JS, Cassar JPh, Wartelle F, Dhulster P, Suhr H: Real time in situ<br />

microscopy for animal cell-concentration monitoring during high<br />

density culture in bioreactor. J Biotechnol 2004, 111:335-343.<br />

2. Joeris K, Frerichs JG, Konstantinov K, Scheper T: In-situ microscopy: Online<br />

process monitoring of mammalian cell cultures. Cytotechnology 2002,<br />

38:129-34.<br />

3. Guez JS, Cassar JPh, Wartelle F, Dhulster P, Suhr H: The viability of Animal<br />

cell Cultures: Can it be Estimated Online by using In Situ Microscopy?<br />

Process Biochem 2010, 45:288-291.<br />

4. Wiedemann P, Egner F, Wiegemann HB, Quintana JC, Storhas W, Guez JS,<br />

Schwiebert C, Suhr H: Advanced in situ microscopy for on-line<br />

monitoring of animal cell culture. Proceedings of the 21st Annual Meeting<br />

of the European Society for Animal Cell Technology (ESACT), Dublin, Ireland,<br />

June 7-10, 2009 Dordrecht Heidelberg London New York: Springer: Jenkins<br />

N, Barron N, Alves P 2011.<br />

Page 111 of 181<br />

5. Wiedemann P, Guez JS, Wiegemann HB, Egner F, Asanza-Maldonado D,<br />

Filipaki M, Wilkesman J, Schwiebert C, Cassar JP, Dhulster P, Suhr H: In<br />

situ microscopic cytometry enables noninvasive viability assessment<br />

of animal cells by measuring entropy states. Biotechnol Bioeng 2011.<br />

P78<br />

Characterization of the human AGE1.HN cell line: a systems biology<br />

approach<br />

Sebastian Scholz 1 , Miriam Luebbecke 2 , Alexander Rath 3 , Eva Schraeder 1 ,<br />

Thomas Rose 4 , Heino Büntemeyer 1 , Thomas Scheper 2 , Udo Reichl 3 ,<br />

Thomas Noll 1*<br />

1 Institute of Cell Culture Technology, University of Bielefeld, Germany;<br />

2 Institute of Technical Chemistry, Leibniz University of Hannover, Germany;<br />

3 Max Planck Insitute for Dynamics of Complex Technical Systems,<br />

Magdeburg, Germany; 4 ProBioGen AG, Berlin, Germany<br />

E-mail: thomas.noll@uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P78<br />

Background: With the emergence of functional genomics approaches in the<br />

past decade, powerful tools for the discovery and understanding of cellular<br />

mechanisms are accessible. The main purpose of those analyses is to<br />

investigate the cell on the level of the transcriptome, proteome and<br />

metabolome to retrieve information on possible limitations for growth or<br />

productivity. In subsequent steps, these limitations could be addressed by<br />

genetic modifications (e.g. overexpression of genes coding for bottle neck<br />

enzymes) or supplementation of the media.<br />

In this work, we present a systems biology approach for the analysis of<br />

the human protein expression cell line AGE1.hn. The focal point of the<br />

analyses was on the central energy metabolism, namely glycolysis, TCA<br />

and oxidative phosphorylation.<br />

In addition to the metabolome analysis transcriptome and proteome data<br />

were obtained. Combining the above mentioned techniques we could<br />

gather valuable insights in the cellular processes of a human protein<br />

production cell line.<br />

Material and methods: The alpha1-antitrypsin expressing AGE1.HN.AAT<br />

cells were cultivated in a 20L STR reactor under controlled conditions<br />

(Temperatur 37°C; pH 7.15; 40% DO).The batch cultivation was carried out in<br />

the protein-free, chemically defined medium 42MAX-UB (Bielefeld<br />

University) with an addition of 5 mM Glutamine.<br />

For the metabolome analysis a targeted LC-MS method was established.<br />

Using a HILIC column for metabolite separation and a Triple-Quad ESI-<br />

MS for the detection, over 50 intracellular metabolites could be<br />

quantified.<br />

Changes in the transcriptome level were detected with a whole genome<br />

DNA microarray (Eurogentec Human HOA 4.3) and further analyzed using<br />

the EMMA software [1]. For the proteome analysis a 2D-DIGE approach<br />

was conducted and >100 differentially expressed protein spots were<br />

analyzed with the DELTA-2D software (DECODON GmbH, Germany). In<br />

subsequent analyses theses spots were identified via nanoLC-MS and<br />

MALDI-MS.<br />

Results: The AGE1.HN.AAT cells showed growth up to a viable cell density<br />

of 5·10 6 cells·mL -1 at day 6 of the cultivation. Total cultivation time was 8<br />

days. The average growth rate during the exponential phase was 0.41 d -1 .<br />

24 hours after inoculation daily sampling for polyomic analyses was<br />

started. Initial concentrations of the extracellular metabolites glucose,<br />

lactate, and pyruvate were 25 mM, 8 mM and 2 mM, respectively.<br />

Both glucose and pyruvate were depleted at day 5 of the cultivation. The<br />

lactate concentration increased to a maximum of 38 mM at day 5.<br />

Subsequent to the maximum concentration, lactate was consumed by the<br />

cells.<br />

Figure 1 shows the changes of the intracellular metabolite concentrations<br />

and gene expression in the TCA cycle during the batch cultivation. Most<br />

of the intracellular metabolites exhibit a concentration time course similar<br />

to glucose.<br />

Following the depletion of pyruvate at day 4 a majority of the TCA<br />

metabolites started to decrease until the end of the cultivation. Yet, the<br />

metabolites were not entirely consumed but remained at low<br />

concentrations. In contrast, malate and a-ketoglutarate concentrations<br />

diminished sharply after the first sampling at day 2 and persisted on these<br />

levels.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P78) Viable Cell Density, extracellular (glucose&lactate) and intracellular TCA metabolite concentrations and corresponding gene<br />

expression profile of a batch cultivation of AGE1.HN cells.<br />

The transcriptome data suggest a strong gene expression for enzymes of the<br />

central metabolism in the first two days of the cultivation, corresponding<br />

with the elevated intracellular glucose and pyruvate concentrations. While<br />

for most genes the expression decreased over time, various genes showed a<br />

high expression throughout the cultivation, notably the malic enzyme,<br />

which is responsible for the conversion of malate into pyruvate under<br />

formation of NADH/NADPH.<br />

Despite the depletion of nutrients feeding glycolysis and the TCA cycle (e.g.<br />

glucose, glutamine) both the adenylate energy charge (AEC) and the<br />

catabolic reduction charge (CRC) (data not shown) remained on consistent<br />

levels.<br />

Conclusion: The presented results indicate a truncated connectivity<br />

between glycolysis and TCA cycle via the conversion of pyruvate to acetyl-<br />

CoA. A recent publication showed similar results for the AGE1.HN cells<br />

using metabolic flux analysis [2]. A possible explanation for the observed<br />

truncation is the relatively abundant gene expression of the PDK4 gene in<br />

the exponential growth phase. The PDK4 enzyme is known to inhibit the<br />

pyruvate dehydrogenase complex thus limiting the conversion of pyruvate<br />

to acetyl-CoA [3]. As a result, a major fraction of the pyruvate is directly<br />

converted into lactate. Thus, a highly inefficient metabolism is favored in<br />

Page 112 of 181<br />

the beginning of the cultivation as long as the levels of glucose and<br />

pyruvate are high enough.<br />

Yet, although the glycolytic flux is directed towards the energetic less<br />

favorable lactate formation, the amount of ATP generated during<br />

glycolysis seems to be sufficient to sustain the energy requirements (AEC,<br />

CRC) of the cell.<br />

In addition with yet unpublished data, these findings offer certain points<br />

of action for further cell line or media optimization. Among others, the<br />

elevated gene expression makes the PDK4 gene a possible target for cell<br />

line engineering.<br />

In conclusion, the integration of polyomics techniques allows a deeper<br />

insight in the metabolism of mammalian cells to identify possible targets<br />

for modifications of cells, media formulation or process strategies.<br />

References<br />

1. Dondrup M: EMMA: a platform for consistent storage and efficient<br />

analysis of microarray data. J Biotech 2003, 106(2-3):135-146.<br />

2. Niklas J, Schräder E, Sandig V, Noll T, Heinzle E: Quantitative<br />

characterization of metabolism and metabolic shifts during growth of<br />

the new human cell line AGE1.HN using time resolved metabolic flux<br />

analysis. Bioprocess. Biosyst. Eng 2010, 533-545.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

3. Sugden MC, Holness MJ: Mechanisms underlying regulation of the<br />

expression and activities of the mammalian pyruvate dehydrogenase<br />

kinases. Arch.Physiol.Biochem2006, 112(3):139-49.<br />

P79<br />

Influence of the nickel-titanium alloy components on biological<br />

functions<br />

Akiko Ogawa 1* , Ryota Akatsuka 1 , Hidekazu Tamauchi 2 , Katsuya Hio 3 ,<br />

Hideyuki Kanematsu 1<br />

1 Suzuka National College of Technology, Suzuka, Japan, 510-0294; 2 Mie<br />

Prefecture Industrial Research Institute Metal Science Branch, Kuwana, Japan,<br />

511-0937; 3 Kitasato University School of Medicine, Sagamihara, Japan, 225-8555<br />

E-mail: ogawa@chem.suzuka-ct.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):P79<br />

Background: Nickel-titanium alloy is applied to biomaterials such dental<br />

wire, stents and so on, because they have outstanding capacities for<br />

corrosion-resistant, adequate mechanical strength and shape-memory [1,2].<br />

On the other hand they have a risk of metal allergy because of leaking nickel<br />

ions from them in aqueous conditions. It is not clear the reason why nickel<br />

causes allergy and what the rate of components is suitable for biomaterial<br />

therefore we have to design biocompatible nickel-titanium alloys based on<br />

experiences. In order to create biological friendly nickel-titanium alloys more<br />

effectively, it is necessary to make the relationship clear between characters<br />

of nickel-titanium alloys and biological reactions. In this study, we<br />

investigated 1) the material properties of nickel-titanium alloys, 2) the effect<br />

of these alloys on cellular function and 3) the allergy test for these alloys.<br />

Materials and methods: Titanium particles were mixed with nickel<br />

particles to become 5%, 15%, 25% or 50% titanium by weight and the<br />

nickel-titanium alloys were made by arc melting. The components of nickeltitanium<br />

alloy were determined by X-ray fluorescence analysis. Nickel plate<br />

was used as a control. Nickel plate and nickel-titanium alloys were cut into<br />

determined surface area by sharing machine then soaked in 70% ethanol<br />

due to sterilize. The sterilized ones were utilized for cellular assay. First, they<br />

were soaked in PBS and heated at 36.5 °C for 25 hours in order to make<br />

extracts. Next, each extract was added to the culture supernatant of MOLT-3<br />

cells (Riken bioresource center cell bank, Japan) and the MOLT-3 cells were<br />

Page 113 of 181<br />

cultured with 10% FBS containing RPMI for 5 days at 36.5 °C in humidified<br />

air containing 5% of CO 2. After that, the viable cell numbers were<br />

determined by the trypan blue exclusion assay and counting in a<br />

hemacytometer under a phase contrast microscope. Both nickel and<br />

titanium concentrations of the extracts were determined by ICP atomic<br />

emission spectrometer (Shimadzu, Japan). Nickel-titanium alloys were<br />

shaped into round columns ( : 10 mm, depth: 5 mm) then sterilized and<br />

transplanted to dorsal subcutaneously of mice. After 28 days, a certain<br />

amount of nickel solution was injected into the auricularis skin of the mice<br />

and the degree of turgid auriculae was measured.<br />

Results: In cellular assay with MOLT-3, the viable cell number of the cells<br />

cultured with the extract of nickel-5% or 50% titanium alloy was significantly<br />

more than that of nickel plate (Figure 1). In allergy test, transplanting nickel-<br />

50% titanium alloy indicated the lower degree of turgid auriculae than<br />

transplanting nickel plate in a mouse but nickel-5% or 15% titanium alloy<br />

did not. About nickel concentration of the extract, nickel-50% titanium alloy<br />

was the lowest and nickel-25% titanium alloy was the highest.<br />

Conclusions: We studied the relationship between characters of nickeltitanium<br />

alloys and biological reactions. Four kinds of nickel-titanium<br />

alloys having different titanium contents were tested. Except nickel-50%<br />

titanium alloy, the nickel concentration of the extract of nickel-titanium<br />

alloy increased with the content rate of titanium. This result suggests that<br />

increasing the content rate of titanium will promote leaking nickel from<br />

nickel-titanium alloys. Otherwise the viable cell number decreased with<br />

the content rate of titanium. The extract of nickel-50% titanium alloy<br />

indicated not only the largest viable cell number among all extracts but<br />

also the lowest nickel concentration. These results indicate that the<br />

amount of nickel would affect cell survival of MOLT-3 cells, and that<br />

nickel concentration of the extracts was not correlated with the titanium<br />

content rates of nickel-titanium alloys but it was done with the structural<br />

states of them. When nickel-titanium alloys were transplanted into mice,<br />

only nickel-50% titanium alloy affected the allergic reaction for the better.<br />

This result suggests that cellular assay will be more sensitive to detect<br />

the material differences of nickel-titanium alloys than animal test.<br />

References<br />

1. Fukushima O, Yoneyama T, Doi H, Hanawa T: Corrosion resistance and<br />

surface characterization of electrolyzed Ti-Ni alloy. Dental Materials<br />

Journal 2006, 25:151-160.<br />

Figure 1(abstract P79) Effect of extract of nickel-titanium alloys on cell survival of MOLT-3 cells. MOLT-3 cells were seeded in 12-well plate at 96,800<br />

cells/well (n =4). Next day, each extract of nickel-titanium alloy or nickel plate was added to the culture supernatant.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

2. DiBari GA: Nickel alloys. The properties of electrodeposited metals and alloys<br />

Florida: The American electroplaters and surface finishers society: Safranek<br />

W H , 2 1986, 327-364.<br />

P80<br />

Metabolomics - a useful tool for prediction of protein production and<br />

processing?<br />

Jennifer Kronthaler 1,2* , Timo Schmidberger 2 , Christine Heel 2<br />

1 2<br />

University of Innsbruck, Innsbruck, Austria; Sandoz Biopharmaceuticals,<br />

Langkampfen, Austria<br />

E-mail: jennifer.kronthaler@sandoz.com<br />

BMC Proceedings 2011, 5(Suppl 8):P80<br />

Introduction: Although CHO cells are widely used as hosts for recombinant<br />

protein production, still only little is known regarding the interaction of<br />

metabolic alterations and protein production and processing. Therefore, a<br />

more sophisticated look into the cellular metabolism might lead to a more<br />

efficient use of the production medium resulting in high quantities of<br />

the protein with the desired product quality. For intracellular metabolite<br />

quantification the sample preparation including quenching of cells is a very<br />

critical step strongly affecting subsequent results. A simple and straightforward<br />

protocol, not needing special equipment, was investigated focusing<br />

on the efficiency of stopping metabolic activities and the potential metabolic<br />

leakage due to losses in membrane integrity. In a next attempt, the<br />

comparison between the producer cell line and the corresponding mock cell<br />

line using targeted mass spectrometry approaches for intra- and extracellular<br />

metabolite quantification was performed. The influence of the increased<br />

protein production capacity and the need of a complex glycosylation<br />

machinery on metabolite profiles was assessed. In order to find a link<br />

between the determined output parameters, detailed data evaluation<br />

including multivariate data analysis tools was implemented.<br />

Materials and methods: CHO producer cell line as well as corresponding<br />

mock cell line (transfected with an empty plasmid) were cultivated as a fedbatch<br />

process in serum-free, chemically defined in-house medium,<br />

comprising insulin. For the experiments 15L bioreactors were used.<br />

Conditions were 36,5°C and ph 6,9. Samplingforsubsequentmetabolite<br />

quantification (intra- as well as extracellular) took place at various time<br />

points throughout cultivation.<br />

Targeted metabolomic analysis was performed for cell culture supernatants<br />

as well as for cell lysates using freeze/thaw cycles after resuspension in<br />

phosphate buffer for extraction. For the quantification of amino acids,<br />

hexose and biogenic amines commercially available AbsoluteIDQ KIT<br />

Figure 1(abstract P80) Intracellular nucleotide sugar concentrations.<br />

Page 114 of 181<br />

plates were used. This fully automated assay was based on PITC<br />

(phenylisothiocyanate)-derivatization in the presence of internal standards<br />

followed by LC-MS/MS detection using a AB SCIEX 4000 QTrap mass<br />

spectrometer with electrospray ionization [1]. For the quantitative analysis<br />

of energy metabolism intermediates (glycolysis, citrate cycle, urea cycle), a<br />

hydrophilic interaction liquid chromatography (HILIC)-ESI-MS/MS method<br />

in highly selective negative MRM detection mode was used. Intracellular<br />

amounts of nucleotides and nucleotide sugars were analyzed by liquid<br />

chromatography-electrospray ionization-mass spectrometry on surfaceconditioned<br />

porous graphitic carbon as described by Pabst et al. [2].<br />

Results: The comparison between a CHO producer cell line and the<br />

corresponding mock cell line enabled the assessment of increased protein<br />

production capacity and the need of a complex glycosylation machinery<br />

influencing metabolite profiles. Multivariate data analysis using a PLS (partial<br />

least square) model on all quantified compounds showed similar progress<br />

throughout fermentation for all four cultivations (producer and mock in<br />

duplicates). Changes between different sampling points, which represent<br />

different stages of cultivation showed the strongest impact. Thus,<br />

differences between tested cell lines have to be evaluated for each phase<br />

individually as no overall alteration is detectable.<br />

Nucleotide sugars which play important key roles within the mammalian<br />

glycosylation pathway showed the major variance between producer and<br />

mock cell line, as shown in figure 1: Early phase represents data within<br />

exponential phase (up to and including day 7) whereas late phase shows<br />

averaged data of day 10 and 12. The increased glycosylation processing in<br />

producer cells seemed to lead to higher intracellular concentrations of<br />

required precursors due to increased stimulation of the overall<br />

glycosylation machinery. Otherwise, an increased consumption by<br />

producer cells should have led to lower concentrations or even to limiting<br />

bottlenecks. The increased amounts of intracellular nucleotide sugars at<br />

late stage for both cell lines were evident, but an adequate explanation<br />

with respect to cell- and process knowledge is still missing.<br />

Moreover, within exponential phase (sampling on day 3-7) specific<br />

consumption/production rates of divers metabolites were different<br />

between producer and mock cell line. Mostly, amino acids were affected.<br />

Mock cells showed an increased energy demand reflected by higher<br />

specific consumption rates. Thus, especially the slightly higher growth rate<br />

of mock cells seemed to be of relevance resulting in a differentiation<br />

between proliferating and producing cells. However, detailed investigation<br />

of impacts of involved pathways on recombinant protein production and<br />

processing is still ongoing.<br />

Conclusion and outlook: Within this study the application of detailed<br />

metabolite data to mammalian process development was evaluated


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

preliminarily. Just marginal differences between producer and mock cells<br />

were detectable. However, further pathway-based investigations are still<br />

ongoing.<br />

References<br />

1. Freepatentsonline.com: 2007 [http://www.freepatentsonline.com/<br />

20070004044.html].<br />

2. Pabst M, Grass J, Fischl R, Leonard R, Jin C, Hinterkorner G, et al: Nucleotide<br />

and nucleotide sugar analysis by liquid chromatography-electrospray<br />

ionization-mass spectrometry on surface-conditioned porous graphitic<br />

carbon. Anal Chem 2010, 82:9782-9788.<br />

P81<br />

Accelerating process development through analysis of cell metabolism<br />

Dirk Müller * , Florian Kirchner, Stefanie Mangin, Klaus Mauch<br />

Insilico Biotechnology AG, D-70563 Stuttgart, Germany<br />

E-mail: dirk.mueller@insilico-biotechnology.com<br />

BMC Proceedings 2011, 5(Suppl 8):P81<br />

Background: Biopharma process development accumulates growing<br />

collections of physicochemical product information and fermentation data,<br />

partly in response to initiatives like Process Analytical Technology (PAT)<br />

and Quality by Design (QbD) backed by regulatory authorities. The key<br />

goal of these initiatives is to ensure robust processes and consistent<br />

product quality based on a scientific and mechanistic understanding of the<br />

product compound itself and of the manufacturing process. Biopharma<br />

companies gather much high-quality data during fermentations including<br />

online measurements and omics data such as transcript and metabolite<br />

measurements. Typically, these data will be stored for documentation<br />

purposes only whereas data evaluation and interpretation lags behind and<br />

often is sporadic at best.<br />

Cellular network models can tap this underused resource for predicting<br />

fermentation outcomes and for analyzing why certain fermentations failed<br />

or succeeded based on a mechanistic representation of cell physiology.<br />

In particular, network models of cell metabolism upgrade metabolomics<br />

data by enabling predictions of cell behavior from concentration time<br />

series of extracellular and – if available – intracellular metabolites. Model<br />

simulations can be used for rapid hypothesis testing, e.g. to evaluate the<br />

impact of changes in feeding on intracellular metabolism, growth, or<br />

product formation. Identifying suitable metabolic target genes for cell line<br />

engineering represents another application area of such models. Here, we<br />

illustrate this approach using the prediction of optimal media compositions<br />

for a Chinese hamster ovary (CHO) cell line employing a genomebased<br />

CHO network model as example.<br />

Methods: The CHO stoichiometric metabolic network was reconstructed<br />

using information from public databases as well as from primary literature<br />

and accounts for the specific amino acid composition and glycoform<br />

structure of the product molecule. In a first step, we applied the network<br />

model to a comprehensive metabolic characterization of the existing<br />

fermentation process. Rates of cellular nutrient uptake, growth, and<br />

product formation in physiologically distinct process phases were<br />

determined from concentration time series of extracellular metabolites<br />

during a fermentation run. These cell-specific rates served to compute<br />

intracellular flux distributions using the CHO network model. Comparing<br />

flux distributions for different process phases provided insight as to when<br />

and where in intracellular metabolism significant changes occur during the<br />

fermentation. This is often not obvious from inspection of concentration<br />

time series alone. Especially for fed-batch processes, multiple feed streams<br />

and volume changes due to pH control and sampling impede<br />

interpretation of raw data. If desired, further information about the usage<br />

of alternative intracellular pathways and in vivo reaction reversibilities can<br />

be obtained from labeling experiments combined with transient 13 C-<br />

Metabolic Flux Analysis [1,2], which is applicable to industrial fed-batch<br />

settings.<br />

Intracellular flux distributions also provide an ideal starting point for process<br />

optimization. Distinct optimal media compositions were computed for<br />

different fermentation phases based on the observed nutrient demand of<br />

the clone inferred from flux distributions. The chosen optimization approach<br />

combines stationary and dynamic model simulations on high-performance<br />

computing clusters. For dynamic simulations, the stoichiometric CHO<br />

network representation was transformed into a kinetic model. Model<br />

parameters were determined using evolutionary strategies and cluster<br />

computing based on the observed metabolite time series and considering<br />

thermodynamic constraints on reaction directionality. Integration of<br />

intracellular metabolite data into this workflow is easy and can further<br />

increase the predictive capabilities of the resulting model. The dynamic<br />

model also comprised a description of the fermenter including feeds and<br />

sampling. In this way, it can be predicted how changes in medium<br />

composition and feed flows impact rates of cell growth, productivity, and<br />

byproduct formation as well as intracellular metabolite profiles. Finally,<br />

media were optimized to maximize final product titer and specific<br />

productivity by varying the concentrations of glucose and individual amino<br />

acids in two continuous feed streams using evolutionary strategies on highperformance<br />

computing clusters.<br />

Results: The resulting optimized media were tested experimentally in a fedbatch<br />

process. The improved feeding resulted in a 50% increase of final<br />

product titer and in an increased integral of viable cells already in the first<br />

iteration (Figure 1). Simultaneously, ammonium release declined markedly. If<br />

desired, the procedure can be repeated to further optimize cellular growth<br />

and/or productivity profiles using data collected from the first evaluation<br />

fermentation as input. Considering replicate fermentations aids in assessing<br />

and improving the robustness of the predicted media compositions, but is<br />

not a prerequisite. The mechanistic model captures stoichiometric couplings<br />

between observed substrate uptake and resulting growth, product synthesis<br />

and byproduct formation. Consequently, the present approach requires<br />

much fewer fermentations runs as input for media optimization compared<br />

Figure 1(abstract P81) Optimized fed-batch media maintained (a) high viable cell counts and (b) resulted in a 50% increase in product titer.<br />

Page 115 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

to standard Design of Experiment (DoE) techniques, thus saving time<br />

and resources. Presently, the prediction focuses on amino acids and carbon<br />

sources, but the extension to further compounds is technically<br />

straightforward.<br />

Conclusions: The combination of metabolomics data and network models<br />

not only improves our quantitative understanding of cell physiology, but<br />

can also support and accelerate multiple steps of rational process<br />

development strategies:<br />

Tailor media compositions to specific clones and curtail the time and<br />

experimental effort required for medium optimization compared to<br />

standard DoE techniques<br />

Identify highly productive and robust clones for scale-up through<br />

comprehensive metabolic characterization during selection at small scales<br />

Devise cell line engineering strategies for overcoming metabolic<br />

bottlenecks in cell growth and product formation by incorporating<br />

intracellular metabolite measurements and other omics data (proteins,<br />

transcripts)<br />

Employ metabolic network models for controlling feed additions at the<br />

production scale.<br />

The above methodology enables biologics manufacturers to add value to<br />

data collected in PAT and QbD initiatives and to harness metabolomic data<br />

for quantitatively predicting and optimizing fermentation outcomes. This<br />

approach is not restricted to CHO cells, but is readily transferable to other<br />

cell lines and also to microbial production strains.<br />

References<br />

1. Schaub J, Mauch K, Reuss M: Metabolic flux analysis in Escherichia coli by<br />

integrating isotopic dynamic and isotopic stationary 13 C labeling data.<br />

Biotechnol Bioeng 2008, 99:1170-1185.<br />

2. Maier K, Hofmann U, Reuss M, Mauch K: Identification of metabolic fluxes<br />

in hepatic cells from transient 13 C-labeling experiments: Part II. Flux<br />

estimation. Biotechnol Bioeng 2008, 100:355-370.<br />

P82<br />

Evaluation of sampling and quenching procedures for the analysis of<br />

intracellular metabolites in CHO suspension cells<br />

Judith Wahrheit * , Jens Niklas, Elmar Heinzle<br />

Biochemical Engineering Institute, Saarland University, 66123 Saarbrücken,<br />

Germany<br />

E-mail: j.wahrheit@mx.uni-saarland.de<br />

BMC Proceedings 2011, 5(Suppl 8):P82<br />

Background: Metabolomics, aiming at the quantification of all extracellular<br />

and intracellular metabolites, is a valuable tool for characterizing,<br />

understanding and manipulating the physiology of mammalian cells. While<br />

extracellular metabolite analysis is well established, required quenching and<br />

extraction procedures for intracellular metabolite analysis in mammalian<br />

suspension cells are not yet routinely available. In this study a simple<br />

sampling and quenching protocol using ice-cold 0.9% saline as quenching<br />

solution [1] was tested on CHO cells. Quenching efficiency, preservation of<br />

cell integrity as well as cell separation and the necessity of washing steps<br />

were evaluated and possible sources of error are discussed.<br />

Materials and methods: The antithrombin-III producing CHO cell line<br />

T-CHO-ATIII was cultivated in serum free CHO-S-SFM II medium (Gibco/<br />

Invitrogen, Carlsbad, CA, USA). Cells were kept in baffled shake flasks<br />

(Corning, Corning, NY, USA) in a shaking incubator (2 in. orbit, Innova 4230,<br />

New Brunswick Scientific, Edison, NJ, USA) at 37°C with a constant 5% (v/v)<br />

CO 2 supply at 185 rpm. Quenching was performed by mixing 5 ml of cell<br />

suspension with 45 ml ice-cold 0.9% ice-cold saline as quenching solution<br />

(QS) [1]. Unless otherwise stated, sampling was done by centrifuging for<br />

1 min at 1000g followed by shock-freezing of cell pellets in a CO 2-acetone<br />

bath. Intracellular metabolites were extracted from freeze-dried cell pellets<br />

using ice-cold 50% acetonitrile [1]. Survival of the cells in the QS at 0°C was<br />

tested after sampling at different relative centrifugal forces (RCF, 1000g –<br />

2000g – 3000g – 4000g). Cell number, viability and cell morphology of cells<br />

re-suspended in QS were checked during incubation at 0°C using an<br />

automated cell counter (Countess, Invitrogen, Karlsruhe, Germany).<br />

Carryover of extracellular metabolites from the culture medium was<br />

investigated without washing and after applying different washing<br />

strategies. Cell pellets were either re-suspended in 50 ml QS or rinsed with<br />

50 ml QS followed by another centrifugation step. Cell numbers and<br />

extracellular metabolites were analysed and compared to the initial sample.<br />

Page 116 of 181<br />

Quenching efficiency at 0°C was investigated by measuring enzyme activity<br />

and by monitoring intracellular metabolite amounts at 0°C. Lactate<br />

dehydrogenase activity was determined in quenched cells (0°C) and in cells<br />

without quenching (37°C) after cell permeabilization with 0.05% Triton-X-100<br />

[2]. Intracellular citrate, a-ketoglutarate, pyruvate, succinate, lactate and<br />

fumarate were quantified in cell extracts after incubation of quenched cell<br />

suspensions at 0°C for 0, 5 or 10 min.<br />

Results: Cell integrity was maintained in ice-cold 0.9% saline for at least<br />

30 min. Centrifugation at 1000g for 1 min led to 50% cell loss during<br />

sampling. Sampling at higher RCF increased cell yield to 65%. However,<br />

sampling at RCF higher than 2000g did not further increase cell yield but<br />

seemed to affect cell viability. Unchanging cell diameter and morphology<br />

before and after quenching and during incubation at 0°C indicated that no<br />

osmotic stress and no biased selection of cells during centrifugation<br />

occurred. Contamination with culture medium was found relatively low<br />

even without any washing steps. Less than 0.25% pyruvate and lactate and<br />

less than 0.1% glucose compared to the amounts measured in the<br />

extracellular medium of the cell suspension were found after quenching.<br />

Citrate found in the initial sample was not detected after quenching in any<br />

of the samples with or without washing. Furthermore, after washing by<br />

rinsing or re-suspending the cell pellets no glucose and less than 0.2%<br />

pyruvate and lactate could be detected. Washing by resuspending did not<br />

yield better results than rinsing the cell pellet but can possibly lead to cell<br />

damage and leakage of intracellular metabolites as indicated by traces of<br />

fumarate detected in the sample. The inclusion of washing steps further<br />

decreases cell yield. Cell loss after rinsing the cell pellet was very low (48%<br />

cell recovery compared to 56% without washing). In contrast, washing by<br />

re-suspending the pellet led to a tremendous cell loss; only 19% of the<br />

initial cell number could be recovered. In quenched cells enzyme activity<br />

was efficiently stopped as shown by comparing lactate dehydrogenase<br />

activity in quenched cells (specific activity 0.33 ± 1.1 pmol/(cell×h)) and<br />

control cells kept at 37°C (15.17 ± 0.19 pmol/(cell×h)). Increasing<br />

concentrations of pyruvate, lactate and fumarate after 10 min incubation<br />

of quenched cell suspensions at 0°C indicate that metabolism is not<br />

completely stopped at 0°C. However, metabolic activity seemed to be<br />

sufficiently slowed down within the first 5 min of incubation where no<br />

significant change of intracellular metabolites was observed.<br />

Conclusions: Ice-cold 0.9% saline proved to be a suitable QS maintaining<br />

cell integrity and cell morphology. Sampling via centrifugation at RCF higher<br />

than 2000g seemed to affect cell viability and should be avoided. By using a<br />

tenfold excess of QS compared to the sample volume rapid cooling of the<br />

sample could be achieved and contamination with culture medium was<br />

found relatively low even without any washing steps. Carryover of<br />

extracellular metabolites can be further reduced by rinsing the cell pellet<br />

with an excess of QS. Washing by re-suspending the cell pellet diminished<br />

cell yield tremendously and can possibly lead to cell damage resulting in<br />

leakage of intracellular metabolites. It is therefore not suitable. In quenched<br />

cells enzyme activity was efficiently halted within the first 5 min of<br />

incubation at 0°C indicating sufficient quenching of metabolic activity for<br />

the analysis of intracellular metabolites with lower turnover. Sampling and<br />

washing should therefore be completed within the first 5 min after<br />

quenching.<br />

References<br />

1. Dietmair S, Timmins NE, Gray PP, Nielsen LK, Krömer JO: Towards<br />

quantitative metabolomics of mammalian cells: Development of a<br />

metabolite extraction protocol. Anal Biochem 2010, 404:155-164.<br />

2. Niklas J, Melnyk A, Yuan Y, Heinzle E: Selective permeabilization for the<br />

high-throughput measurement of compartmented enzyme activities in<br />

mammalian cells. Anal Biochem 2011, doi:10.1016/j.ab.2011.05.039.<br />

P83<br />

Increasing productivity of hybridoma cell lines by sorting by side<br />

scattering light<br />

Daniel Landgrebe * , Cornelia Kasper, Thomas Scheper<br />

Institute for Technical Chemistry, Leibniz University of Hannover, 30167<br />

Hannover, Germany<br />

BMC Proceedings 2011, 5(Suppl 8):P83<br />

Introduction: The side scattering light of a mammalian cell is caused,<br />

among other things, by the membranes of cell organelles. We suggest that<br />

cells with a high side scatter contain a large amount of mitochondria and a


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

large endoplasmatic reticulum. In a simple approach we separate cells with<br />

a high side scatter via fluorescence activated cell sorting (FACS) to create<br />

sub populations with a higher productivity. We obtain cells with a high<br />

amount of mitochondria and a large endoplasmatic reticulum which could<br />

cause strong energy metabolism and high protein productivity. The<br />

advantage of this technique is that no staining dye or complex procedure<br />

is needed to reach the goal of increasing the productivity of a cell line.<br />

Materials and methods: For the sorting procedure the SC-71 murine<br />

hybridoma cell line (DSMZ, Braunschweig) is used. The cells produce an IgG 1<br />

specific for rat myosin. Cultivation of the cells before and after the sorting<br />

steps was performed at the same conditions. The cells were grown in 100<br />

ml Erlenmeyer shacking flasks with 20 ml medium (DMEM (Sigma-Aldrich,<br />

Steinheim),10% FCS, 4mM glutamine, 1% Penicillin/Streptomycin solution<br />

(PAA, Cölbe) ; conditions: 37°, 5% CO 2, 110 rpm) with an inoculation density<br />

of 0.45*10 6 cells/ml. During the cultivation samples were taken twice a day<br />

for the acquisition of the cell number and offline analysis (product<br />

concentration via ELISA (Roche, Mannheim), glucose and lactate via YSI 2700<br />

Biochemistry Analyzer (Yellow Springs Instruments) and amino acids via<br />

HPLC (Agilent Technologies, Waldbronn)). The product concentration is used<br />

to calculate the specific productivity rate of the cell lines as described by<br />

Brezinsky et al. [1].<br />

For each sorting 5*10 6 cells were taken from the exponential phase of a<br />

preparatory culture. After a washing step with PBS the cells were stained<br />

with Propidium iodide (5 µl [1 mg/ml] (Sigma-Aldrich, Steinheim)). Only<br />

Propidium iodide negatives cells were sorted to exclude dead cells.<br />

Aggregates were excluded by analyzing the pulse heights and their pulse<br />

integrals. For the separation cells with the maximal 3% of the side scatter<br />

were sorted into a 6 well plate with 1ml of DMEM using a FACSVantage (BD<br />

Biosciences, Heidelberg) equipped with pulse processing. After an expansion<br />

time of 4 weeks the cells were used to repeat the procedure and to<br />

investigate growth and productivity qualities. The procedure were repeated<br />

three times to generate altogether four cell lines, one initial cell line and<br />

three sub cell lines (named population I – III according to the repeated<br />

sorting procedures).<br />

Subsequent flow cytometric analyses were used to verify the constancy of<br />

the sorting effect. Therefore the four populations were cultivated for two<br />

month. Passages were performed by medium changes every third or fourth<br />

day. Samples were taken after the twenties passage. 0.5*10 6 cells of each<br />

cell line were washed with PBS and the side scatter was analyzed using an<br />

Epics XL-MCL flow cytometer (Beckman Coulter, Krefeld).<br />

Figure 1(abstract P83) Specific productivity rates of initial population and sub populations.<br />

Page 117 of 181<br />

Results: The calculation of the specific productivity rates show that a higher<br />

specific productivity is achieved earlier in the sub populations than in the<br />

initial population (Fig. 1). Nevertheless the total productivity is the same. Sub<br />

populations I and III show a maximal specific productivity already after 36 h<br />

(resp. after 60 h for sub population II). Compared with the initial population<br />

which reaches their max. after 72 h, it is a considerable increase of a<br />

important process parameter. The sub populations show also an increased<br />

cell density of 10 to 30%. This effect is retrograding in the later sub<br />

populations. The cytometric analyses of the long term cultivation show that<br />

the separated cells have kept an increased side scatter. This effect is stable<br />

for at least 2 month of cultivation.<br />

Conclusions: The results suggest that the sub populations have a modified<br />

metabolism. This could be a result of the accumulation of mitochondria. To<br />

confirm this further experiments are necessary. These experiments should<br />

include staining of mitochondria and endoplasmatic reticulum and<br />

subsequent flow cytometric analysis to determine the amount of these<br />

organelles. Further investigations for the consumption of the main<br />

metabolites glucose and glutamate and the production of lactate could<br />

explain the changes in the growth behavior and productivity of the cells.<br />

Acknowledgement: This work was performed within the project<br />

“SysLogics: Systems biology of cell culture for biologics” and is founded<br />

by the Bundesministerium für Bildung und Forschung (FKZ 0315275F)<br />

Reference<br />

1. Brezinsky SC, Chiang GG, Szilvasi A, Mohan S, Shapiro RI, MacLean A, Sisk W,<br />

Thill G: A simple method for enriching populations of transfected CHO cells<br />

for cells of higher specific productivity. J Immunol Methods 2003, 277:141-155.<br />

P84<br />

Compartmentation and channelling of metabolites in the human cell<br />

line AGE1.HN®<br />

Jens Niklas 1* , Volker Sandig 2 , Elmar Heinzle 1<br />

1 Biochemical Engineering Institute, Saarland University, 66123 Saarbrücken,<br />

Germany; 2 ProBioGen AG, 13086 Berlin, Germany<br />

E-mail: j.niklas@mx.uni-saarland.de<br />

BMC Proceedings 2011, 5(Suppl 8):P84<br />

Background: A thorough knowledge of the metabolism and its<br />

compartmentation in mammalian cells is desirable to enable rational<br />

design and optimization of producing cell lines and production processes


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

for biopharmaceuticals. In this study we focused on acquiring a detailed<br />

understanding of metabolite channelling and the metabolic flux<br />

distribution during overflow metabolism in the human cell line AGE1.HN®<br />

(ProBioGen AG). This metabolic phenotype characterized by energy spilling<br />

as well as waste product formation is commonly observed in the<br />

beginning of the cultivation [1]. 13 C tracer experiments and 13 Cflux<br />

analysis as applied in this investigation represent methods offering indepth<br />

insights into cellular physiology [2,3].<br />

Materials and methods:<br />

Cultivation and analysis: The human neuronal cell line AGE1.HN®<br />

(ProBioGen AG, Berlin, Germany) was used. Cultivations were carried out in<br />

shake flasks (Corning, NY, USA) or bioreactor filter tubes (TPP, Trasadingen,<br />

Switzerland). 13 C-labeling experiments using the tracers [1,2- 13 C 2] glucose ,<br />

[U- 13 C 5]glutamine,[U- 13 C 3] alanine, [1- 13 C 1] lactate (Cambridge Isotope<br />

Laboratories, Andover, MA, USA) and [U- 13 C 6] glucose (Euriso-Top,<br />

Saarbrücken, Germany) were conducted. Extracellular metabolites were<br />

quantified using different HPLC methods [4,5]. 13 C-labeling of metabolites<br />

was analysed using GC-MS [6]. Carbon mass isotopomers were determined<br />

from the analyte mass isotopomer distribution [7].<br />

Carbon atom transition model: A carbon atom transition model was set<br />

up using the Kyoto Encyclopedia of Genes and Genomes (www.kegg.com)<br />

pathway database for Homo sapiens.<br />

13 C metabolic flux analysis: Fluxes were estimated using the method of<br />

Yang et al. [8] applying Matlab R2008 (The Mathworks, Natick, MA, USA).<br />

Results: Experiments applying 13 C-labelled glucose, glutamine, alanine and<br />

lactate tracers were carried out to identify active pathways and channelling<br />

of metabolite carbons in the central metabolism of AGE1.HN®. It was<br />

observed that almost 80% of glucose consumed was detected in lactate.<br />

Smaller amounts were channeled to alanine (5%) and serine (3%). Glucose<br />

carbons were additionally entering the tricarboxylic acid (TCA) cycle which<br />

can be deduced from an increase in fractional labelling of glutamate and<br />

proline in glucose tracer experiments. Reflux from TCA cycle metabolites to<br />

glycolytic metabolites was also detected since labelling in lactate as well as<br />

alanine was measured when using [U- 13 C 5] glutamine as tracer. Decrease in<br />

labelled extracellular lactate as well as an increase in labelled extracellular<br />

alanine as observed in the lactate tracer experiment shows that lactate was<br />

not only produced but also taken up during overflow metabolism.<br />

Furthermore, the lactate and alanine tracer experiments indicated that<br />

alanine and lactate and subsequently the pyruvate pools used for their<br />

synthesis are connected. Alanine taken up was mainly transaminated<br />

entering the cytosolic pyruvate pool, converted to lactate and secreted.<br />

Fluxes were calculated for the growth phase between day 1 and day 4 of<br />

the cultivation in which the cells exhibit overflow metabolism characterized<br />

by production of waste metabolites [1].<br />

For flux calculation extracellular and anabolic rates as well as the labelling<br />

information stored in extracellular lactate resulting from three different<br />

parallel tracer experiments using [1,2- 13 C 2]glucose,[U- 13 C 6]glucoseand<br />

[U- 13 C 5] glutamine was included. In theory one could also use the labelling<br />

information stored in the building blocks of the cellular macromolecules,<br />

namely proteins, carbohydrates, lipids or nucleic acids as it is widely applied<br />

in microbial 13 C flux analysis. However, the time that is needed to<br />

incorporate the labelling in these molecules as well as the presence of huge<br />

amounts of unlabelled species caused by high seeded cell densities needed<br />

in mammalian cell culture processes is a huge disadvantage. Another<br />

possibility would be using the labelling patterns of intracellular metabolites.<br />

This requires, however, sampling methods that are still not well established<br />

in mammalian suspension cell culture. A main problem represents hereby<br />

the quenching procedure that remains still a huge challenge for suspension<br />

cell culture and it cannot be guaranteed that the measured labelling or<br />

intracellular concentration is really representing intracellular values not<br />

corrupted with extracellular species. The labelling of extracellular amino<br />

acids, which could also be used theoretically, is however not directly<br />

representing the labelling of intracellular species. This is caused by the fact<br />

that the amino acids are provided in the medium and the reversibility of<br />

production and uptake of these and their conversion dilutes the labelling<br />

which is then not representing the labelling of metabolites in central<br />

metabolism of the cell. This could only be solved by detailed knowledge<br />

and modelling of reaction reversibilities. In this study solely lactate labelling<br />

was therefore included in the metabolic flux estimation since it is not<br />

present in the beginning of the cultivation representing at steady state<br />

directly the labelling of cytosolic pyruvate which is probably the most<br />

important metabolic hub in the cell.<br />

Page 118 of 181<br />

It was observed that the flux through the oxidative branch of the pentose<br />

phosphate pathway was very low being around 2% of the glycolytic flux<br />

which might be compensated in AGE1.HN® by a high flux through malic<br />

enzyme additionally producing NADPH. The cytosolic pyruvate pool was fed<br />

mainly by the glycolytic flux, however, pyruvate taken up was also<br />

significantly contributing to intracellular pyruvate. Cytosolic pyruvate was<br />

almost exclusively converted to lactate and alanine and secreted. Flux<br />

between cytosolic oxaloacetate and cytosolic pyruvate was found to be<br />

reversible with similar fluxes in both directions. Pyruvate transport between<br />

mitochondria and cytosol was very low. Mitochondrial oxaloacetate pool<br />

was fed by cytosolic oxaloacetate. The flux from mitochondrial oxaloacetate<br />

to mitochondrial pyruvate was relatively high. The tricarboxylic acid cycle<br />

was fed mainly via a-ketoglutarate and oxaloacetate.<br />

Conclusions: In order to improve metabolic efficiency in AGE1.HN®<br />

engineering strategies focussing on improved metabolite transfer between<br />

cytosolic and mitochondrial pyruvate pools could be applied accompanied<br />

by improved control and targeted reduction of substrate availability and<br />

uptake. The 13 C flux analysis method that was applied in this study allows a<br />

detailed determination of metabolic fluxes in the central metabolism of<br />

mammalian cells and can thus be applied for studying related biological<br />

questions. The presented data is an important step for an improved system<br />

level understanding of the AGE1.HN cell line.<br />

References<br />

1. Niklas J, Schräder E, Sandig V, Noll T, Heinzle E: Quantitative<br />

characterization of metabolism and metabolic shifts during growth of<br />

the new human cell line AGE1.HN using time resolved metabolic flux<br />

analysis. Bioprocess Biosyst Eng 2011, 34(5):533-545.<br />

2. Niklas J, Schneider K, Heinzle E: Metabolic flux analysis in eukaryotes. Curr<br />

Opin Biotechnol 2010, 21(1):63-9.<br />

3. Niklas J, Heinzle E: Metabolic Flux Analysis in Systems Biology of<br />

Mammalian Cells. Adv Biochem Eng Biotechnol 2011, DOI: 10.1007/<br />

10_2011_99. [Epub ahead of print]..<br />

4. Niklas J, Noor F, Heinzle E: Effects of drugs in subtoxic concentrations on<br />

the metabolic fluxes in human hepatoma cell line Hep G2. Toxicol Appl<br />

Pharmacol 2009, 240(3):327-336.<br />

5. Krömer JO, Fritz M, Heinzle E, Wittmann C: In vivo quantification of<br />

intracellular amino acids and intermediates of the methionine pathway<br />

in Corynebacterium glutamicum. Anal Biochem 2005, 340(1):171-173.<br />

6. Wittmann C, Hans M, Heinzle E: In vivo analysis of intracellular amino acid<br />

labelings by GC/MS. Anal Biochem 2002, 307(2):379-382.<br />

7. Yang TH, Bolten CJ, Coppi MV, Sun J, Heinzle E: Numerical bias estimation<br />

for mass spectrometric mass isotopomer analysis. Anal Biochem 2009,<br />

388(2):192-203.<br />

8. Yang TH, Frick O, Heinzle E: Hybrid optimization for 13C metabolic flux<br />

analysis using systems parametrized by compactification. BMC Syst Biol<br />

2008, 26:2-29.<br />

P85<br />

Producer vs. parental cell – metabolic changes and burden upon a1antitrypsin<br />

production in AGE1.HN®<br />

Jens Niklas 1* , Christian Priesnitz 1 , Volker Sandig 2 , Thomas Rose 2 ,<br />

Elmar Heinzle 1<br />

1 Biochemical Engineering Institute, Saarland University, 66123 Saarbrücken,<br />

Germany; 2 ProBioGen AG, 13086 Berlin, Germany<br />

E-mail: j.niklas@mx.uni-saarland.de<br />

BMC Proceedings 2011, 5(Suppl 8):P85<br />

Background: The human designer cell line AGE1.HN® represents a<br />

promising production system for biopharmaceuticals, particularly for those<br />

needing human-type post-translational modifications [1,2]. For further<br />

rational improvement of the cell line and the cultivation process a detailed<br />

understanding of the metabolism and metabolic changes during<br />

glycoprotein production is desirable. Metabolism and metabolic burden<br />

upon production of a 1-antitrypsin (A1AT) were analyzed by comparing<br />

parental cells and a derived clone, AGE1.HN.AAT. The questions addressed<br />

were (i), which changes occur in cell growth and metabolism upon A1AT<br />

production and (ii), what are specific cellular properties distinguishing the<br />

producer clone AGE1.HN.AAT from the parental cell population.<br />

Materials and methods:<br />

Cultivation and analysis: Cultivations of the human neuronal cell line<br />

AGE1.HN® (ProBioGen AG, Berlin, Germany) were carried out in shake


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

flasks (Corning, NY, USA). Extracellular metabolites were quantified using<br />

different HPLC methods [3,4]. A1AT was quantified using an activity assay.<br />

Identification of extracellular proteins was done using MALDI-ToF/ToF MS<br />

(Applied Biosystems). Biomass components were quantified using classical<br />

biochemical methods.<br />

Model for A1AT production: Model for A1AT production was set up<br />

using the Kyoto Encyclopedia of Genes and Genomes (http://www.kegg.<br />

com) pathway database for Homo sapiens.<br />

Metabolic flux analysis: Fluxes were estimated using standard methods<br />

[1,5] by incorporating extracellular and anabolic rates using Matlab<br />

R2007b (The Mathworks, Natick, MA, USA).<br />

Results: Cell growth was similar in the first 5 days. After 5 days AGE1.HN.AAT<br />

cells were growing less than the parental cell line and the viability dropped<br />

faster. After 2 days dry weight was slightly higher in the parental cell line.<br />

A1AT production was low in the first 2 days and then increasing up to a final<br />

concentration of ~0.4 g/l in the end of the cultivation. The total cell mass<br />

was increasing similarly in the first 4 days. After 4 days AGE1.HN.AAT cell<br />

mass remained constant whereas the extracellular protein concentration was<br />

increasing. This was different in the AGE1.HN parental cells where the cell<br />

mass was still increasing after 4 days and the increase in total extracellular<br />

protein mass was less. Intracellular protein concentration was decreasing in<br />

AGE1.HN.AAT after 4 days. Specific cellular proteins might be degraded upon<br />

nutrient deprivation to maintain the production of the recombinant<br />

glycoprotein A1AT. This was clearly different in the parental cell line.<br />

Fractions of the biomass constituents RNA, lipid and phosphatidylcholine<br />

Figure 1(abstract P85) Schematic presentation of the glycoprotein production process in mammalian cells.<br />

Page 119 of 181<br />

were increased in AGE1.HN.AAT. The total mass of the analyzed components<br />

in the end of the cultivation was similar in the cultivations of both cell lines<br />

being around 2.5 g/l. The final A1AT concentration which was 0.4 g/l was<br />

~30% of the total protein in the culture.<br />

Similar time courses for most extracellular metabolites were observed.<br />

Glycine and glutamate production was higher in AGE1.HN.AAT whereas<br />

uptake of arginine and aspartate as well as alanine production were lower.<br />

Glutamine was consumed in the cultivations of both cell lines at ~4 days<br />

which resulted also in reduced growth. Especially the increase in glycine and<br />

glutamate production points to differences in C1 metabolism and cellular<br />

nucleotide demand.<br />

The differences that can be seen in the metabolism upon production of the<br />

glycoprotein were explained by using an appropriate model of the anabolic<br />

demand needed to produce active A1AT. The whole cellular production<br />

process of a glycoprotein includes its expression (transcription), synthesis of<br />

the amino acid chain (translation), posttranslational modification in<br />

endoplasmic reticulum (ER) and Golgi apparatus (Golgi) and secretion of the<br />

mature protein (Figure 1). Metabolite demand for A1AT production was<br />

finally simulated. These results indicate that one could expect an increase in<br />

the production of glutamate and glycine with increasing intracellular<br />

nucleotide demand which was observed for AGE1.HN.AAT; this is caused by<br />

differences in cellular composition of the producer, partly originating from<br />

recombinant A1AT production.<br />

The differences that were found in the biomass composition between both<br />

cell lines were included in a metabolic network model and intracellular


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

metabolic fluxes were calculated for both cell lines. Fluxes in central energy<br />

metabolism (glyolysis, TCA cycle) were similar. Differences were observed<br />

in C1 and glutamate metabolism including changes in activities of several<br />

transaminases. The observed changes in metabolic flux reflect the anabolic<br />

differences between producer and parental cells.<br />

Conclusions: This improved understanding of the metabolism and cellular<br />

changes during glycoprotein production supports the identification of<br />

targets for further improvement of (i) cell line, e.g., by genetic modifications<br />

and (ii) cultivation process, e.g., by improved feeding strategies. The<br />

presented data indicate that nucleotide and lipid metabolism might be<br />

interesting targets for further engineering of the AGE1.HN® cell line.<br />

References<br />

1. Niklas J, Schräder E, Sandig V, Noll T, Heinzle E: Quantitative<br />

characterization of metabolism and metabolic shifts during growth of<br />

the new human cell line AGE1.HN using time resolved metabolic flux<br />

analysis. Bioprocess Biosyst Eng 2011, 34(5):533-545.<br />

2. Blanchard V, Liu X, Eigel S, Kaup M, Rieck S, Janciauskiene S, Sandig V,<br />

Marx U, Walden P, Tauber R, Berger M: N-glycosylation and biological<br />

activity of recombinant human alpha1-antitrypsin expressed in a novel<br />

human neuronal cell line. Biotechnol Bioeng 2011, 108:2118-2128.<br />

3. Niklas J, Noor F, Heinzle E: Effects of drugs in subtoxic concentrations on<br />

the metabolic fluxes in human hepatoma cell line Hep G2. Toxicol Appl<br />

Pharmacol 2009, 240(3):327-336.<br />

4. Krömer JO, Fritz M, Heinzle E, Wittmann C: In vivo quantification of<br />

intracellular amino acids and intermediates of the methionine pathway<br />

in Corynebacterium glutamicum. Anal Biochem 2005, 340(1):171-173.<br />

5. Niklas J, Heinzle E: Metabolic Flux Analysis in Systems Biology of<br />

Mammalian Cells. Adv Biochem Eng Biotechnol 2011, DOI: 10.1007/<br />

10_2011_99. [Epub ahead of print].<br />

P86<br />

Analysis of the mitochondrial subproteome of the human cell line<br />

AGE1.HN – a contribution to a systems biology approach<br />

Eva Schräder 1* , Sebastian Scholz 1 , Volker Sandig 2 , Raimund Hoffrogge 1 ,<br />

Thomas Noll 1<br />

1 Institute for Cell Culture Technology, University of Bielefeld, Bielefeld,<br />

Germany; 2 ProBioGen AG, Berlin, Germany<br />

E-mail: esc@zellkult.techfak.uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P86<br />

Background: In Systems Biology approaches a good amount of reliable data<br />

is essential for effective modelling of distinct biological processes. Hence the<br />

focus of the SysLogics-Project was on modelling of the central metabolism<br />

using different functional genomic techniques. For displaying the involved<br />

proteins, it is crucial to enrich them using subproteomic fractions. As many<br />

enzymes being involved in central metabolism are located in the<br />

Page 120 of 181<br />

mitochondria, we investigated the expression dynamics of mitochondrial<br />

proteins during batch cultivations of human AGE1.HN cells (ProBioGen,<br />

Berlin, Germany).<br />

Materials and methods:<br />

Cultivation and isolation of mitochondria: Cultivations were performed<br />

as batch-processes with chemically-defined and animal-component-free<br />

media 42-MAX-UB (Teutocell, Bielefeld, Germany) with addition of 5 mM<br />

glutamine. The processes were performed in a 20 L-stainless steel vessel<br />

(Sartorius-Stedim, Germany). After disintegration of cells by addition of glass<br />

beads (0,1 mm diameter) and vortexing, the isolation of mitochondria was<br />

performed with sucrose density-centrifugation in an ultracentrifuge<br />

(Beckman Coulter, Krefeld, Germany). Protein extraction from mitochondria<br />

was performed with lysis-buffer, which contains Tris-HCl, NaCl, EDTA, SDS,<br />

PMSF and NP40.<br />

Proteomics: The first dimension of 2D-electrophoresis was performed with<br />

Ettan IPGPhor3-system*, using pH 3-11 (NL) IPG-strips*. For generating the<br />

mitochondrial master-map, six gels with 0.45 mg protein extract were<br />

prepared. For DIGE-approaches 0.15 mg were applied, each sample with<br />

four replicates. Second dimension was accomplished with Ettan Dalt six<br />

Electrophoresis System*. 2DE-gels with DIGE-staining were processed with<br />

Ettan Dige Imager*. Software evaluation was implemented with Delta2D<br />

(Decodon, Germany).<br />

After isolation and tryptic digest protein-identification was performed with<br />

MS/MS using MALDI-ToF/ToF (UltrafleXtreme, Bruker, Germany), followed<br />

by database-searching.<br />

*(GE Healthcare, Sweden)<br />

Results: For proteomic analyses, mitochondria were isolated and<br />

successfully confirmed by staining with Rhodamine 123, a mitochondriaspecific<br />

fluorescent dye. 2DE-gels of mitochondrial fraction led to 519<br />

separated spots. 280 proteins could be reliably identified, out of which more<br />

than 50% are exclusively mitochondrial. This 2D-proteome map is our basis<br />

for approaches of analysing different protein expression in mitochondria.<br />

Two standardised batch cultivations in a 20 L-stirred tank stainless steel<br />

vessel were carried out with daily sampling for metabolomics, transcriptomics,<br />

metabolic flux and proteomics, in order to generate reliable and<br />

comparable samples. Batch cultivation in 20 L-scale of AGE1.HN.AAT shows<br />

cell densities and duration as expected. Glucose- and lactate-concentrations<br />

developed also as estimated in standard batch culture (see fig. 1a+b).<br />

Subproteomic DIGE-analysis of mitochondrial proteins in those 20 Lbatch<br />

cultivations resulted in 114 proteins that were significantly<br />

differently expressed in the first and 206 proteins in the second process<br />

during cultivation time (p < 0.05, factorial Anova). More than 40<br />

proteins were found to be significantly regulated in both experiments.<br />

DIGE-approaches of mitochondrial fraction of both batch cultivations led<br />

to expression profiles of identified proteins. Four examples of expression<br />

profiles of typical proteins of TCA and respiratory chain are shown<br />

below in table 1.<br />

Figure 1(abstract P86) a (left): VCD (solid) and viability (dashed) during batch process 1 (open squares) and 2 (closed triangles) Fig. 1b (right):<br />

Concentrations of glucose (solid) and lactate (dashed) during batch process 1 (open squares) and 2 (closed triangles).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P86) Examples of expression profiles of TCA and respiratory chain enzymes. Expression normalised<br />

on sample point 1<br />

ZE1 (2,6 d) ZE1 (4,8 d) ZE1 (6,5 d) ZE2 (2,9 d) ZE2 (4,2 d) ZE2 (5,3 d) ZE2 (6,9 d)<br />

Pyruvate dehydrogenase E1a 1,53 1,6 0,83 1,51 1,56 1,09 0,87<br />

L-lactate dehydrogenase B chain 0,95 0,87 5,18 0,98 1,53 2,4 3,26<br />

Dihydrolipoamide S-succinyltransferase 2,37 2,18 1,2 2,06 1,92 1,73 0,89<br />

Electron transfer flavoprotein alpha 2,57 2,24 1,38 2,29 1,88 1,99 1,45<br />

Apart from these examples it was possible to observe important<br />

biological processes with our 2DE-approach, as for instance:<br />

- tricarboxylic acid cycle<br />

- respiratory chain<br />

- anti-apoptosis and apoptosis<br />

- signal transduction<br />

- chaperone/protein folding and processing<br />

- fatty acid-/lipid-metabolism<br />

- amino acid-degradation<br />

- inhibition of DNA-synthesis<br />

- ketone-metabolism<br />

- protein-biosynthesis<br />

- redox-regulation<br />

- ubiquinone-biosynthesis<br />

Hence, we see this data as a valuable contribution to the Systems Biology<br />

approach for modelling central metabolism events.<br />

Conclusions: Reproducible methods for isolation of mitochondrial proteins<br />

from cultured cells for further analysis, such as proteomic 2DE-approaches,<br />

have been established. DIGE-analysis of mitochondrial subproteome of<br />

AGE1.HN.AAT cells result in the identification of more than 280 identified<br />

proteins, of which more than 40 are regulated during batch cultivation.<br />

Proteomics data in conjunction with the other “omics"-results offer a<br />

promising basis for characterisation of central metabolism during Systems<br />

Biology approaches.<br />

Acknowledgments<br />

This work is part of the SysLogics Project: Systems biology of cell culture for<br />

biologics, founded by German Ministry for Education and Research (BMBF).<br />

P87<br />

Characterisation of cultivation of the human cell line AGE1.HN.AAT<br />

Eva Schräder 1* , Sebastian Scholz 1 , Jens Niklas 5 , Alexander Rath 2 ,<br />

Oscar Platas Barradas 3 , Uwe Jandt 3 , Volker Sandig 5 , Thomas Rose 5 ,<br />

Ralf Pörtner 3 , Udo Reichl 2 , An-Ping Zeng 3 , Elmar Heinzle 4 , Thomas Noll 1<br />

1 Institute for Cell Culture Technology, University of Bielefeld, Germany; 2 Max<br />

Planck Institute for Dynamics of Complex Technical Systems, Magdeburg,<br />

Germany; 3 Institute for Bioprocess and Biosystems Engineering, Hamburg<br />

University of Technology, Germany; 4 Biochemical Engineering Institute,<br />

Saarland University, Saarbruecken, Germany; 5 ProBioGen AG, Berlin, Germany<br />

E-mail: esc@zellkult.techfak.uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P87<br />

Background: Human cell lines are an interesting alternative to CHO cells for<br />

the production of recombinant proteins and monoclonal antibodies,<br />

Page 121 of 181<br />

because of their ability to produce genuine human posttranslational<br />

modifications. The human cell line AGE1.HN.AAT (ProBioGen, Berlin,<br />

Germany), that originated from human neural precursor tissue, has been<br />

adapted to serum-free conditions and cultivated in many different systems.<br />

Here we present our results using this cell line in a scale-up of batch<br />

cultivation from 50 mL vented polypropylene tube on a shaking platform,<br />

polycarbonate shakeflask (cultivation volume from 50 mL up to 300 mL), a 2<br />

L-glass vessel stirred tank reactor and a 20 L-stainless steel stirred tank<br />

reactor (both Sartorius Stedim, Goettingen, Germany).<br />

Materials and methods: Cultivations were performed with chemicallydefined<br />

and animal-component-free media 42-MAX-UB (Teutocell, Bielefeld,<br />

Germany). Batch-cultivations were performed in 50 mL-bioreactor tubes (TPP,<br />

Switzerland) shakeflasks (Corning Life Sciences, Netherlands), 2 L-glass vessel<br />

and 20 L-stainless steel vessel (both Sartorius-Stedim, Germany). Chemostatcultivation<br />

was done in 0.5 L-bioreactor (DASGIP, Juelich, Germany) with a<br />

media exchange rate of 7 mL/h. As a further cultivation system a dialysisreactor<br />

(Bioengineering, Wald, Switzerland) was established, with a 1.3 L cellcontaining<br />

inner chamber and a 4 L media reservoir in the outer chamber,<br />

separated by a semipermeable dialysis membrane.<br />

Results:<br />

Proliferation in controlled and uncontrolled systems: The AGE1.HN.<br />

AAT cells show similar growth in different unregulated vessels and<br />

culture volumes. The used systems ranged from 50 mL-bioreactor tubes<br />

with a culture volume of up to 18 mL, 125 mL-shakeflasks with a culture<br />

volume of up to 50 mL- to 250 mL-shakeflasks, in which a culture volume<br />

of 100 mL can be used (Results shown in figure 1a).<br />

Cultivation in 2 L-glass vessel is as well feasible for batch process as well as<br />

preculture for 20 L-vessel. Cultivation at a 20 L-scale resulted in delayed cell<br />

growth but did not affect the final cell concentration (refer to figure 1b).<br />

AGE1.HN.AAT cells show a strong growth-coupled productivity as shown in<br />

figure 1c.<br />

No difference between 2 L- and 20 L-vessel concerning spec. productivity<br />

and spec. growth rate were observed. A scale-up of cultivation-volume in<br />

batch process is definitely possible.<br />

Cultivations with other systems: Cultivation of AGE1.HN.AAT cells in<br />

dialysis-reactor is also possible and batch cultivation without mediaexchange<br />

in the outer chamber showed maximum cell density up to 1.6 E7<br />

viable cells per milliliter in the inner chamber after eight days of cultivation.<br />

Chemostat-cultivation in 0.5 L-glass vessel shows constant cell density at<br />

2.5 E6 cells/mL for more than 14 days.<br />

Conclusions: Cultivation of AGE1.HN cell line is possible in different<br />

regulated and unregulated systems and at different scales. Cell specific<br />

productivity and titer of the AGE1.HN.AAT producer cell line depend mainly<br />

on cell growth. The cells are easily scalable from 2 to 20 L batch cultivation<br />

Figure 1(abstract P87) 1.1 (left): viable cell density and viability during shake flask batch-cultivation. open squares: bioreactor tube, open circles: 125 mLshakeflask,<br />

open diamonds: 250 mL-shakeflask. 1.2 (middle): viable cell density and viability during bioreactor batch-cultivation. open squares: 2 L-glass<br />

vessel, open circles: 20 L-stainless steel reactor. 1.3 (right): specific growth rate μ of bioreactor cultivation vs. corresponding specific productivity qP. open<br />

diamonds: 20 L- stainless steel reactor, open squares: 2 L-glass vessel.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

in STR. Other cultivation strategies have been established successfully (incl.<br />

chemostat and dialysis-bioreactor) documenting the potential of the AGE1.<br />

HN cell line.<br />

Acknowlegments: The work presented was part of SysLogics (Systems<br />

biology of cell culture for biologics), which is funded by German Federal<br />

Ministry of Education and Research. Cultivation of 125 mL shakeflasks and<br />

bioreactor tubes were done in Saarbruecken, dialysis cultivation in Hamburg,<br />

chemostat-process was done in Magdeburg, while 250 mL shakeflask, 2<br />

L-glass vessel and 20 L-stainless steel bioreactor-cultivation were performed<br />

in Bielefeld.<br />

P88<br />

Strategies in umbilical cord-derived mesenchymal stem cells expansion:<br />

influence of oxygen, culture medium and cell separation<br />

Antonina Lavrentieva 1* , Tim Hatlapatka 1 , Ramona Winkler 1 , Ralf Hass 2 ,<br />

Cornelia Kasper 2<br />

1 Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5,<br />

D-30167 Hannover; 2 Hannover Medical School, Klinik für Frauenheilkunde<br />

und Geburtshilfe, AG Biochemie und Tumorbiologie, Carl-Neuberg-Str. 1,<br />

D-30625 Hannover<br />

E-mail: kasper@iftc.uni-hannover.de<br />

BMC Proceedings 2011, 5(Suppl 8):P88<br />

Background: Mesenchymal stem cells (MSC) from different sources attract<br />

tremendous interest in cell-based therapies for their ability to differentiate<br />

into different cell lineages. MSC are already used in clinical trials as cell<br />

therapy [1]. In comparison to other “classical” sources of MSC (e.g. bone<br />

marrow and adipose tissue), the umbilical cord (UC) matrix has great<br />

potential, since there are no ethical limitations, risks for the donor or<br />

problems with variety of donor age [2]. However, a large numbers of cells<br />

(about 10 6 cells per 1 kg body weight) are required which may still limit the<br />

implant preparation for clinical applications. Thus, strategies for the<br />

expansion of MSC must be well defined and controlled conditions need to<br />

be developed and established for reproducible production of cells under<br />

GMP conform conditions. Conventional in vitro cell cultivation is carried out<br />

under ambient oxygen concentration (21% of O2) whichisdefinedas<br />

“normoxic”. MSCin vivo usually are not exposed to such a high concentration<br />

of oxygen. In our work we studied the influence of oxygen<br />

concentration on the long-term as well as glucose and oxygen concentration<br />

on the short-term cultivation and expansion of UC-MSC.<br />

Materials and methods: UC-MSC were isolated from whole human<br />

umbilical cords using an explant culture approach and characterised as<br />

described earlier [3,4]. For the long-term cultivation, MSC from the same<br />

donor were isolated and subsequently cultivated in two different oxygen<br />

Page 122 of 181<br />

concentrations (5% and 21%). After isolation, cells were seeded at a density<br />

of 2000 cells/cm 2 in 25cm 2 cell culture flasks (Corning, Germany) and subcultivated<br />

every 3-4 days over 25 passages. Cell numbers were estimated at<br />

the end of each passage and cumulative population doublings were<br />

calculated for each culture conditions. MSC were cultivated in aMEM<br />

containing 1 g/l glucose (Biochrom, Germany), 10% allogenic human serum<br />

(provided by the Institute of Transfusion Medicine, Medical University<br />

Hannover, Germany) and 50 µg/ml gentamicin (PAA Laboratories GmbH). To<br />

reveal the influence of glucose concentration on the proliferation capacities<br />

of UC-MSC under different oxygen concentrations, cells were seeded in 6well<br />

plates (Sarstedt, Germany) at a density of 700 cells/cm 2 in aMEM (1 g/l<br />

glucose) and DMEM (4.5 g/l glucose) and cultivated over 7 days until full<br />

confluency was reached. Cell number and viability in all experiments were<br />

determined by trypan blue exclusion (n=4).<br />

Counterflow centrifugal elutriation (CCE) was performed for the separation<br />

of the cells according to their physical size (Beckmann J6-MC with the JE-<br />

5.0 rotor; 5 ml-standard elutriation chamber, Beckman Coulter, Germany).<br />

Small-sized MSC were separated from the primary heterogeneous<br />

population. After cell separation, proliferation activity of the cells was<br />

estimated as described above.<br />

Results: Long-term cultivation of UC-MSC under hypoxia revealed higher<br />

proliferation activity of the cells without changes in morphology when<br />

compared to ambient (21%) oxygen concentration (Fig 1A).<br />

High glucose concentration inhibited cell growth under ambient oxygen<br />

concentration. Hypoxic conditions, however, significantly increased<br />

proliferation of UC-MSC both, in high- and low-glucose culture medium<br />

(Fig.1B).<br />

Conclusions: Application of MSC in regenerative medicine as cell<br />

suspensions or as a part of tissue engineered implant requires large amount<br />

of cells of high quality. MSC expansion under physiological conditions allows<br />

obtaining higher cell numbers in a shorter period of time. Also the glucose<br />

concentration in medium should be close to that in vivo. Highamountsof<br />

glucose inhibit cell proliferation in ambient oxygen concentrations. In<br />

hypoxic conditions MSC proliferate better and retain their ability to<br />

differentiate. We conclude that GMP-conform cultivation of MSC with<br />

allogenic human serum under physiological oxygen concentrations as well<br />

as isolation of actively dividing cells may increase the efficiency of the cell<br />

expansion for further therapeutic applications.<br />

Acknowledgements<br />

This work was supported by the state of Lower Saxony, DFG Project<br />

“GMP-Model Lab Tissue Engineering”, BMWi Project KF22501101SB9<br />

“Frozen Cells”<br />

References<br />

1. Ma T: Mesenchymal stem cells: From bench to bedside. World J Stem<br />

Cells 2:13-17.<br />

Figure 1(abstract P88) (A) Influence of different oxygen concentrations (5 % and 21%) on the long-term UC-MSC cultivation: cells were counted at the<br />

end of each passage and cumulative population doublings were calculated. (B) Influence of glucose concentration on the short-term proliferation of the<br />

MSC: cell growth in high-glucose medium is inhibited under 21% oxygen.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

2. Fong CY, Richards M, Manasi N, Biswas A, Bongso A: Comparative growth<br />

behaviour and characterization of stem cells from human Wharton’s<br />

jelly. Reprod Biomed Online 2007, 15:708-718.<br />

3. Majore I, Moretti P, Stahl F, Hass R, Kasper C: Growth and differentiation<br />

properties of mesenchymal stromal cell populations derived from whole<br />

human umbilical cord. Stem Cell Rev 7:17-31.<br />

4. Lavrentieva A, Majore I, Kasper C, Hass R: Effects of hypoxic culture<br />

conditions on umbilical cord-derived human mesenchymal stem cells.<br />

Cell Commun Signal 8:18.<br />

P89<br />

Abstract withdrawn<br />

BMC Proceedings 2011, 5(Suppl 8):P89<br />

Abstract withdrawn<br />

P90<br />

Abstract withdrawn<br />

BMC Proceedings 2011, 5(Suppl 8):P90<br />

Abstract withdrawn<br />

P91<br />

Discerning key parameters influencing high productivity and quality<br />

through recognition of patterns in process data<br />

Huong Le 1† , Marlene Castro-Melchor 1† , Christian Hakemeyer 2 , Christine Jung 2 ,<br />

Berthold Szperalski 2 , George Karypis 3 , Wei-Shou Hu 1*<br />

1 Department of Chemical Engineering and Materials Science, University of<br />

Minnesota, Minneapolis, MN 55455, USA; 2 Roche Diagnostics GmbH, 82377<br />

Penzberg, Germany; 3 Department of Computer Science and Engineering,<br />

University of Minnesota, Minneapolis, MN 55455, USA<br />

E-mail: wshu@umn.edu<br />

BMC Proceedings 2011, 5(Suppl 8):P91<br />

Page 123 of 181<br />

Background: The adoption of Quality by Design (QbD) approach to<br />

biologics manufacturing requires fundamental understanding of complex<br />

relationship between the quality of the product, especially critical quality<br />

attributes (CQAs), and various parameters of the manufacturing process<br />

[1]. This can be approached through multivariate analysis of historical cell<br />

culture bioprocess data [2]. In this study, process parameters and raw<br />

materials data obtained from 51 runs with final titer varying from 0.8 to<br />

2.0 units and Gal0 glycan ranging from 47.5 to 67.5% was investigated.<br />

The aim was to discover prominent patterns which may cause the spread<br />

of final process outcome.<br />

Materials and methods: Offline and online data were processed using<br />

linear interpolation and a moving window average method, respectively as<br />

described previously [3]. Data from the 1,000 L scale was organized into six<br />

cumulative datasets corresponding to days 3, 6, 8, 10, 13, and 15. Euclidean<br />

distance for each process parameter between all pairs of runs was<br />

calculated and normalized to 0-1. The similarity measure was determined<br />

using exponential transformation of the negative of the corresponding<br />

distance, and organized into a matrix form. The overall similarity matrix was<br />

computed as the weighted combination of all individual similarity matrices.<br />

The weight of each reflects how well it correlates to the deviation in final<br />

process outcome. A support vector regression (SVR) model was constructed<br />

using the overall similarity of all process parameters to predict the final<br />

product titer and glycosylation profiles for each cumulative dataset.<br />

Prediction accuracy was assessed using the Pearson’s correlation coefficient<br />

(r) between the predicted and the actual values.<br />

Results: Final recombinant antibody concentration (final titer) was predicted<br />

with reasonable accuracy using process data at 1,000 L scale. At up to day 3,<br />

online and offline data can be indicative of the final titer with an accuracy of<br />

r = 46%. Inclusion of data at up to day 6 improved the prediction accuracy<br />

markedly to 80%. A modest increase in SVR models predictability was<br />

observed when data from day 8 and day 10 was incorporated with r = 83%<br />

and 87%, respectively. Online and offline data from days 13 and 15 of the<br />

1,000 L biorectors improved model predictability further to 90%, and 92%,<br />

respectively.<br />

Critical process parameters with significant contribution to SVR model<br />

predictability were weighted using a non-linear Spearman’s correlation<br />

coefficient between its similarity for all pairs of runs and the deviation in<br />

their final titer. Parameters with weights (w) greater than 0.2 included<br />

Figure 1(abstract P91) Process runs in three dimensional space of Gal0, Gal1, and Gal2. Low-titer runs are colored in red, and high-titer runs are in blue.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

stirrerspeed,VCD,glucose,LDH,ammonia,andlactate.Eachbears<br />

significant contribution to prediction of the final titer at different time<br />

periods at the 1,000 L scale. Stirrer speed (w = 0.464) appeared to be<br />

critical throughout whereas VCD (w = 0.445) and LDH (w = 0.300)<br />

became important only after day 3. The contribution of glucose (w =<br />

0.416) and ammonia (w = 0.297) began at day 6, followed by titer values<br />

(w = 0.765) at day 8 and lactate (w = 249) at day 10.<br />

Similar results were obtained when Gal0 was used as the objective<br />

function for the SVR models in place of the final titer. A marked increase in<br />

prediction accuracy was also observed when data from day 6 was included<br />

compared to day 3 (from 61% to 85%). After a modest increase to 88% at<br />

day 8, almost no further improvement was made using data from later<br />

days (10, 13, and 15).<br />

Parameters with high correlation to Gal0 content profile were stirrer speed,<br />

VCD, glucose, LDH, ammonia, CO 2, lactate, temperature, and viability.<br />

Among those parameters, six were in common to when final titer was used<br />

as the objective function. Three parameters that were critical to prediction<br />

of Gal0 but not final titer included CO 2, temperature, and viability. The time<br />

periods in which each parameter had significant correlation to Gal0 content<br />

also varies. VCD (w = 0.536), stirrer speed (w = 0.517), and CO 2 (w = 0.354)<br />

were critical from the beginning of the 1,000 L scale. The contribution of<br />

LDH (w = 0.417), temperature (w = 0.236), and lactate (w = 0.293) became<br />

important at day 3. Day 6 marked the emergence of titer values at previous<br />

time points (w = 0.475), glucose (w = 0.424), and ammonia (w = 0.397),<br />

followed by viability (w = 0.226) at day 10.<br />

As final titer and Gal0 content were predicted with similar accuracy using<br />

similar parameters, a possible relationship between them was explored.<br />

Furthermore, other measures of glycosylation profiles such as NG, Gal1,<br />

and Gal2 that are possibly relevant to product quality [4] were also<br />

included in the analysis. k-means clustering (k = 2) was performed to<br />

separate runs into two clusters using each of these measures. Regardless<br />

of the measure, the two resulting clusters are reasonably well separated in<br />

final titer. Cluster 1 mostly corresponds to high final titer whereas cluster 2<br />

to low final titer. In a three dimensional space of Gal0, Gal1, and Gal2,<br />

process runs are also clustered according to their final titer Figure 1). Thus<br />

this observation further confirms an intrinsic correlation between product<br />

quantity and product quality.<br />

Conclusions: A strong correlation between productivity (final titer) and<br />

product quality (Gal0) was observed. Each was predicted with similarly high<br />

accuracy using support vector regression models built upon process data<br />

from the 1,000 L bioreactors. Predictability increased significantly when data<br />

up to day 6 was analyzed as compared to day 3. Prediction accuracy<br />

continued to increase with additional data inclusion but at a slower rate.<br />

Several parameters contributed significantly to the deviation of product<br />

quantity and quality across runs, including stirrer speed, LDH, lactate,<br />

glucose, and VCD. Among those, stirrer speed and VCD appeared to exert<br />

the most critical impact on final process outcome from early stages of the<br />

1,000 L scale. This approach represents an important step towards<br />

understanding process characteristics for enhanced process robustness, and<br />

thus contributes to the advance of bio-manufacturing.<br />

Acknowledgement: The authors would like to thank the Minnesota<br />

Supercomputing Institute (MSI) for computational support.<br />

References<br />

1. Rathore AS, Winkle H: Quality by design for biopharmaceuticals. Nat<br />

Biotech 2009, 27:26-34.<br />

2. Charaniya S, Hu W-S, Karypis G: Mining bioprocess data: opportunities<br />

and challenges. Trends in Biotechnology 2008, 26:690-699.<br />

3. Charaniya S, Le H, Rangwala H, Mills K, Johnson K, Karypis G, Hu W-S:<br />

Mining manufacturing data for discovery of high productivity process<br />

characteristics. Journal of Biotechnology 2010, 147:186-197.<br />

4. Hossler P, Khattak SF, Li ZJ: Optimal and consistent protein glycosylation<br />

in mammalian cell culture. Glycobiology 2009, 19:936-949.<br />

P92<br />

Proteomic and metabolomic characterization of CHO DP-12 cell lines<br />

with different high passage histories<br />

Tim Beckmann * , Tobias Thüte, Christoph Heinrich, Heino Büntemeyer,<br />

Thomas Noll<br />

Institute of Cell Culture Technology, Bielefeld University, 33615 Bielefeld, Germany<br />

E-mail: tbe@zellkult.techfak.uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P92<br />

Page 124 of 181<br />

Background: For industrial pharmaceutical protein production fast<br />

growing, high producing and robust cell lines are required. To select more<br />

pH shift permissive and fast growing sub-populations, the CHO DP-12<br />

(ATCC clone #1934) cell line, an anti-IL8 antibody producing CHO K1<br />

(DHFR - ) clone, was continuously subcultured at high viability (>90%) for<br />

more than four hundred days in shaking flasks using a chemically defined<br />

medium. During this long-term cultivation there was a repeated shift in pH<br />

and most robust and fast growing cells became accumulated [1]. Cell<br />

samples were cryopreserved at four different time points, after 21, 95, 165<br />

and 420 days (in the following named sub-populations (SP)). The effects of<br />

long-term passaging before cryopreservation correlating with an increase<br />

in specific growth rate as well as changes in product formation and<br />

metabolism were examined in parallel bench-top bioreactor cultivation of<br />

SP21, SP95, SP165 and SP420 sub-populations. During exponential growth<br />

phase samples were taken for the analyses of differences in intracellular<br />

metabolites and protein expression (Please consider article “Growth<br />

characterization of CHO DP-12 cell lines with different high-passage<br />

histories” by Heinrich et al. in this issue for a detailed discussion of longterm<br />

cultivation, changes in specific growth rate, product formation and<br />

metabolic shifts).<br />

Material and methods: The CHO DP-12 cell line was cultivated in<br />

chemically defined, animal component-free medium TC 42 (TeutoCell AG)<br />

with addition of 8 mM glutamine and 200 nM methotrexate. For first steps<br />

of suspension adaption PowerCHO-2 (Lonza AG) medium was used. Parallel<br />

bioreactor cultivations were performed in 2 L bench-top vessels with an<br />

initial working volume of 1.2 L. Cultivation parameters were closed-loop<br />

controlled and set to 37 °C, pH 7.1 and 40% DO of air saturation. Cell<br />

counting and determination of viability was performed using a CEDEX<br />

system (Innovatis-Roche AG). Samples for metabolome and proteome<br />

analysis were taken from parallel bioreactor cultivations of the four subpopulations<br />

during exponential growth phase. Changes in the protein<br />

expression were analyzed by differential two-dimensional gel<br />

electrophoresis (GE Healthcare) with four technical replicates inclusive dye<br />

swap, respectively. Image processing and statistical evaluation was carried<br />

out with Delta 2D 4.2 software (Decodon GmbH). All differentially expressed<br />

protein spots were successfully identified using an ultrafleXtreme MALDI-<br />

TOF-TOF (Bruker Daltonics). For the analysis of intracellular metabolites an<br />

in-house developed fast-filtration quenching procedure was used to<br />

generate samples for GC-MS and LC-MS measurements [2]. For GC-MS<br />

analyses keto- and aldehyde functions were converted into their oxime<br />

derivatives using methoxyamine hydrochloride. Other relevant functions like<br />

amines, carboxylic acids or hydroxyls were masked with trimethylsilyl groups<br />

resulting in volatile derivatives. The nucleotide concentrations were<br />

determined by a HILIC-MS method using a MonoChrom diol column<br />

(Varian).<br />

Results: Based on the expression of 1377 detected proteins spots the four<br />

sub-populations could be clearly separated from each other in a principle<br />

component analysis. An ANOVA analysis (a ≤ 0.05, false significant<br />

proportion ≤ 0.01) revealed 43 protein spots with significantly different<br />

abundance. Hierarchical clustering and examination of fold-changes of<br />

significantly different expressed proteins showed that the quantity of<br />

different proteins and the degree of change increased with the number of<br />

passages before cryopreservation. Between SP21 and SP95 only three protein<br />

spots were detected with an at least two-fold change. For SP196 and SP420<br />

sevenand41spotsshowedaratiowithanabsolutevalueoftwoin<br />

comparison with SP21, respectively. In a follow up analysis the correlation of<br />

protein expression and the number of passages was examined. A template<br />

matching approach (R ≥ 0.98) [3] revealed 23 proteins whose abundance was<br />

linearly correlated with the number of days before cryopreserving the cells.<br />

Among these spots anti-stress proteins, candidates involved in protein<br />

folding, glycolytic enzymes and also proteins participating in transcription<br />

regulation, mRNA processing, cytoskeleton formation, protein biosynthesis as<br />

well as folate and purine metabolism were identified. In addition, there was a<br />

set of 10 unique proteins which showed an inverted correlation with the<br />

number of passages. The results from proteomic analysis indicate that the<br />

four subpopulations not only differ in protein abundances directly related to<br />

cell growth, but also show differences in diverse aspects of cellular protein<br />

expression.<br />

The analysis of intracellular metabolites revealed a positive correlation<br />

between the uptake rates of extracellular metabolites and their<br />

intracellular pool size. Interestingly, this trend is not fulfilled for a couple of<br />

the investigated metabolites: For example, the aspartate pool increases


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

while the uptake of extracellular aspartate decreases although it is not at<br />

limiting concentrations. This indicates that the replenishment by<br />

intracellular pathways is favored over the uptake of extracellular substrate<br />

in this case. Furthermore, an increase of the adenine as well as guanine<br />

energy charge was observed for an increasing number of passages. Both,<br />

adenosine- and guanosine-5’-triphosphate, are necessary for anabolic<br />

reactions as well as protein synthesis. They may also play a role in<br />

apoptosis by inhibiting the formation of the apoptosome [4] and therefore<br />

might be one factor for the observed increase in specific growth rate.<br />

By merging the proteomic findings with the measurements of intracellular<br />

metabolite pools we gained a better understanding of factors which let a<br />

production cell line grow faster and be more robust against pH shifts. This<br />

example shows that the analysis of protein expression combined with<br />

measurements of intracellular pool sizes may give additional hints for cell<br />

line, process and media development in addition to classical approaches.<br />

References<br />

1. Heinrich C, Wolf T, Kropp C, Northoff S, Noll T: Growth characterization of<br />

CHO DP-12 cell lines with different high-passage histories. BMC<br />

Proceedings 2011, 5(Suppl 8):P29.<br />

2. Volmer M, Northoff S, Scholz S, Thute T, Buntemeyer H, Noll T: Fast<br />

filtration for metabolome sampling of suspended animal cells. Biotechnol<br />

Lett 2011, 33:495-502.<br />

3. Pavlidis P: Using ANOVA for gene selection from microarray studies of<br />

the nervous system. Methods 2003, 31:282-289.<br />

4. Chandra D, Bratton SB, Person MD, Tian Y, Martin AG, Ayres M,<br />

Fearnhead HO, Gandhi V, Tang DG: Intracellular nucleotides act as critical<br />

prosurvival factors by binding to cytochrome C and inhibiting<br />

apoptosome. Cell 2006, 125:1333-1346.<br />

P93<br />

A method for metabolomic sampling of suspended animal cells using<br />

fast filtration<br />

Martin Volmer, Julia Gettmann * , Sebastian Scholz, Heino Büntemeyer,<br />

Thomas Noll<br />

Institute of Cell Culture Technology, Bielefeld University, D-33615 Bielefeld,<br />

Germany<br />

E-mail: Julia.Gettmann@uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P93<br />

Background: For intracellular metabolomic analyses it is of utmost<br />

importance to rapidly stop the organism’s metabolism during sampling. This<br />

Page 125 of 181<br />

is to avoid false data due to residual enzymatic activity during sampling.<br />

Unlike for bacteria and yeast, there is not one generally approved protocol<br />

for metabolome sampling of suspended animal cells. A number of sampling<br />

procedures have been developed and described in literature but have not<br />

been compared in depth.<br />

Here we describe a sophisticated sampling method for metabolome<br />

analysis of suspended animal cells using a fast filtration protocol. The main<br />

requirements for the fast filtration method were to reduce the time for<br />

quenching and cell-medium separation while reducing cell disruption to a<br />

minimum.<br />

Additionally, the fast filtration method is compared to the other sampling<br />

methods described in literature.<br />

Methods: CHO DP12 cells were cultured in shaker flasks and 2 L Bioreactors<br />

to generate sample cell suspensions for the experiments.<br />

The different quenching methods were used according to the literature.<br />

Methods tested included sampling with fast filtration [1], sampling in cold<br />

saline solution [2], sampling in cold methanol/ammonium bicarbonate<br />

(AMBIC) [3], and sampling with a microstructure heat exchanger [4]. As a<br />

reference sampling of cells using centrifugation without dedicated<br />

quenching was used.<br />

For 13 C-labeling experiments the cells were transferred to saline solution<br />

prior to sampling. 13 C-labeled glucose was added to the cells followed by<br />

immediate sampling. The ratio of labeled and unlabeled glycolysis<br />

metabolites was then determined.<br />

LDH and ATP release from the cells was measured using plate tests. The<br />

intracellular energy metabolism was investigated using HILIC columns on<br />

a LC-MS system.<br />

Results: The cell disruption during sampling was investigated by<br />

measurement of LDH release from the cells. It was low at 5% and 2.5% for<br />

fast filtration and centrifugation, respectively. No influence of the cell<br />

disruption on the measured intracellular metabolite levels was observed.<br />

The intracellular amino acid content per cell was constant for filters loaded<br />

with up to 6x10 7 cells. Thus, indicating a good extraction efficiency with the<br />

tested cell numbers. Furthermore this suggests that no residual medium<br />

components remained on the filter.<br />

The adenylate concentrations were measured in extracts from all tested<br />

sampling methods. These showed good correlation of ATP concentrations<br />

for all sampling methods tested. The same was true for ADP and AMP<br />

concentrations. Thus, fortifying the observation of a good extraction<br />

efficiency of cells on filters.<br />

The adenylate energy charge (AEC) was used as an indicator of quenching<br />

efficiency for the comparison of the proposed method with those from<br />

Figure 1(abstract P93) Percentage of 13 C-labeled hexose 6-phosphate after feeding the cells with 13 C-glucose immediately prior to sampling (N=4).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

literature. A significant advantage over sampling using a microstructure<br />

heat exchanger or a simple centrifugation protocol was observed.<br />

Comparison to quenching in cold methanol with ammonium bicarbonate<br />

and cold saline solution exhibited AEC values similar to quenching with<br />

fast filtration. Furthermore, experiments with isotope labeled glucose<br />

(Figure 1) exhibited only small amounts of labeled glycolysis intermediates<br />

in extracts of samples taken with fast filtration and cold methanol/AMBIC<br />

compared to saline solution quenching and centrifugation. This is most<br />

likely due to the lower temperature when sampling with methanol/AMBIC<br />

and the shorter overall sampling time when using fast filtration.<br />

Metabolite leakage of the two methods with the best quenching efficiency<br />

was done by investigating the ATP concentration in spent washing<br />

solutions after sampling. Sampling with methanol showed ATP leakage of<br />

up to 14% whereas no leakage of ATP was observed when sampling with<br />

the fast filtration method.<br />

Conclusion: The results show that the fast filtration method is well<br />

applicable for metabolome sampling of suspended animal cells. The two<br />

major concerns with this method were that medium components remained<br />

with the cells after filtration and that extraction of metabolites from the filter<br />

would not be efficient [2]. Both of which could be disproved in this study.<br />

The comparison of the different sampling methods described in literature<br />

showed that fast filtration and quenching in cold methanol/AMBIC offer the<br />

best quenching efficiency. The drawback of sampling using methanol is the<br />

metabolite leakage caused by the contact of methanol with the cell<br />

membrane. This leaves the fast filtration method as the best suited method<br />

for reliable metabolome sampling for suspended animal cells.<br />

References<br />

1. Volmer M, Northoff S, Scholz S, Thüte T, Büntemeyer H, Noll T: Fast<br />

filtration for metabolome sampling of suspended animal cells. Biotechnol<br />

Lett 2011, 33:495-502.<br />

2. Dietmair S, Timmins NE, Gray PP, Nielsen LK, Krömer JO: Towards<br />

quantative metabolomics of mammalian cells: development of a<br />

metabolite extraction protocol. Anal Biochem 2010, 404:155-164.<br />

3. Sellick CA, Hansen R, Maqsood AR, Dunn WB, Stephens GM, Goodacre R,<br />

Dickson AJ: Effective quenching processes for physiologically valid<br />

Page 126 of 181<br />

metabolite profiling of suspension cultured mammalian cells. Anal Chem<br />

2009, 81:174-183.<br />

4. Wiendahl C, Brandner JJ, Küppers C, Luo B, Schygulla U, Noll T, Oldiges M:<br />

A microstructure heat exchanger for quenching the metabolism of<br />

mammalian cells. Chem Eng Technol 2007, 30:322-328.<br />

P94<br />

Analysis of glycolytic flux as a rapid screen to identify low lactate<br />

producing CHO cell lines with desirable monoclonal antibody yield and<br />

glycan profile<br />

Rachel Legmann 1* , Julie Melito 1 , Ilana Belzer 2 , David Ferrick 1<br />

1 2<br />

Seahorse Bioscience, N. Billerica, MA, USA; ProCognia, Ashdod, IL, USA<br />

E-mail: rlegmann@seahorsebio.com<br />

BMC Proceedings 2011, 5(Suppl 8):P94<br />

Background: In CHO cell lines currently selected for the production of<br />

recombinant antibody, approximately 80% of the metabolized glucose is<br />

converted into lactic acid. These cells with a glycolytic phenotype exhibit<br />

significantly higher rates of proton production (extracellular acidification<br />

rate, ECAR) from lactate production than cells using oxidative phosphorylation<br />

(oxygen consumption rate, OCR). Therefore, shifts in the cell’s<br />

metabolism can be detected conveniently and dynamically through<br />

simultaneous detection of ECAR and OCR. Such measurements can<br />

characterize the metabolic programming of individual cell types and<br />

forecast the quality potential of their produced glycoproteins. In this study,<br />

we utilized an XF96 analyzer to measure glycolysis and mitochondrial<br />

respiration simultaneously, and in real-time. This allows one to determine<br />

the response of these two pathways to ATP demand, and indirectly,<br />

biosynthetic needs. A rapid screen was performed to determine the desired<br />

lactic acid production by exposing the cells to alternate sources of<br />

substrates, such as galactose or fructose. Specific metrics of the study<br />

included cell growth, product yield, and glycan profile. Higher titer, viable<br />

cell density, and viability along with glycol-similarity were observed for<br />

galactose and glutamine feeding strategies during the production phase.<br />

Figure 1(abstract P94) Seahorse XF analyzer measure the two ATP generating pathways of the cell. The cell is consuming oxygen, producing CO 2 and<br />

excreting (H+) protons that acidify the media.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

We believe this rapid, cell based metabolic screen that is label-free and noninvasive<br />

can be used to identify low lactic acid CHO mAb cell producers in<br />

both batch and fed-batch systems. This selection is accomplished without<br />

compromising clone productivity and product quality.<br />

Material and methods: Suspended CHO cells producing a recombinant<br />

IgG monoclonal antibody (MAb) were maintained in high glucose serum<br />

free chemically-defined (CD) CHO supplemented with 0.2μM methotrexate<br />

(MTX). The low-buffered DMEM assay medium was used for XF96 ECAR<br />

screening and CD CHO medium was used for fed-batch flasks for lactic acid<br />

screening. Mitochondrial function and cellular bioenergetics were measured<br />

in intact CHO_mAb using a Seahorse Bioscience extracellular flux analyzer<br />

(XF96) as described previously (1, 3) and demonstrated in Figure 1. The<br />

sensor cartridge is embedded with 96 dual florescent biosensors (O and H+).<br />

Each sensor cartridge is also equipped delivery ports for injecting agents<br />

during an assay. Cells were maintained in a 5% CO2 incubator at 37 °C<br />

and 1 h before the experiment, cells were washed and incubated in<br />

nonbuffered (without sodium carbonate) DMEM (sugar-free) pH 7.4, at 37 °C<br />

in a non-CO2 incubator. Real time cellular ECAR and OCR were measured<br />

simultaneously before and after the substrate source injection. The<br />

metabolic rate of the cell population was measured repeatedly over about<br />

30 min. Lactic acid concentrations in the flasks were measured after<br />

different carbon sources were added to the cells after 5 days of growth and<br />

glucose depletion during the production phase using a NOVA BioProfile<br />

100 Plus. MAb concentrations in samples from flasks were quantified using<br />

Octet QK instrument with protein A biosensors. Glycan profile in crude<br />

harvest samples from flasks were quantified using lectin based array by<br />

Procognia Glycoscope instrument described previously(2).<br />

The illustration demonstrates how the XF96 measures changes in the<br />

microenvironment (3μl) surrounding live cells in a 96-well microplate. The<br />

O2 and proton concentration is determined by a quenching reaction of a<br />

fluorophore specific for O2 (blue) or protons (red) that is embedded into<br />

a single polymer martix on the biosensor cartridge.<br />

Results: Mammalian cells in culture have inefficient glucose metabolism<br />

where most of the glucose consumed is converted into lactate, while<br />

very little is oxidized in the tricarboxylic acid cycle (TCA).One of the<br />

interpretation for this phenomenon of high glycolysis rate even under<br />

fully aerobic condition is that lactate generation from pyruvate may be<br />

used by these cells as a way to re-equilibrate their redox potential. The<br />

high glycolysis rates would generate NADH at high rates, which should<br />

be reduced to NAD in order to keep this pathway functional. In Figure 2<br />

(A), metabolic analyses from this study demonstrate that different carbon<br />

and nitrogen sources added during the product production phase can<br />

promote or diminish aerobic glycolysis as indicated by ECAR. The data<br />

show that ECAR correlates well with glycolysis driving lactic acid synthesis<br />

in the recombinant CHO_mAb cells. The highest lactic acid concentration<br />

and ECAR were obtained when glucose was present in the medium as<br />

thesolecarbonsource.ThelowestlacticacidandECARwereobtained<br />

when galacose or fructose was present in the media during the<br />

glycoprotein production phase. Therefore, adding galactose or fructose<br />

when glucose becomes depleted at the end of the growth phase would<br />

Page 127 of 181<br />

provide an alternative carbon source to drive low-waste lactic acid<br />

production. The rapid method developed in this study identified the<br />

optimum carbon and nitrogen source that achieved less than 77% proton<br />

production compared to glucose while maintaining optimal and<br />

consistent protein glycosylation. Metabolic shift to aerobic glycolysis was<br />

observed when glucose or mannose was injected into the CHO_mAb cell<br />

cultures as shown in Figure 2B. XF analyses demonstrated that cells<br />

consuming glucose or mannose are divertedtowardglycolysisevenin<br />

the presence of sufficient levels of dissolved oxygen. Good correlation<br />

was observed between ECAR and accumulation of lactic acid in<br />

Erlenmeyer flasks (Figure 2C).<br />

Conclusions: In this study, we developed and validated a rapid assay,<br />

employing the XF96 analyzer, to screen for low ECAR producing cells to<br />

predict low lactic acid production. The experiments show that there is a<br />

good agreement between ECAR and lactic acid and therefore ECAR can<br />

be rapidly measured as an index of glycolytic activity and serve as an<br />

accurate surrogate of lactate production. The data show that alternating<br />

carbon sugars during the production phase ,such as galactose and<br />

fructose, can help to control glycolysis and, therefore, reduce the lactate<br />

accumulation in mammalian cell cultures without compromising on<br />

recombinant glycoprotein productivity and desired glycoform profile.<br />

References<br />

1. Ferrick DA, Neilson A, Beeson C: Advances in measuring cellular<br />

bioenergetics using extracellular flux. Drug Discovery Today 2008, 13(5-<br />

6):268-274.<br />

2. Rosenfeld R, Bangio H, Gerwig GJ, Rosenberg R, Aloni R, Cohen Y, Amor Y,<br />

Plaschkes I, Kamerling JP, Maya RB: A lectin array-based methodology for<br />

the analysis of protein glycosylation. J Biochem Biophys Methods 2007,<br />

70(3):415-426.<br />

3. Wu M, Neilson A, Swift AL, Moran R, Tamagnine J, Parslow D, Armistead S,<br />

Lemire K, Orrell J, Teich J, Chomicz S, Ferrick DA: Multiparameter<br />

metabolic analysis reveals a close link between attenuated<br />

mitochondrial bioenergetic function and enhanced glycolysis<br />

dependency in human tumor cells. Am J Physiol Cell Physiol 2007, 292(1):<br />

C125-36.<br />

P95<br />

Influence of cell specific productivity on product quality<br />

Ruchika Srivastava, Lavanya Rao, Kriti Shukla, Sunaina Prabhu,<br />

Saravanan Desan, Dinesh Baskar * , Ankur Bhatnagar, Anuj Goel, Harish Iyer<br />

Cell Culture Lab, Biocon Limited, Bangalore, India<br />

E-mail: Dinesh.baskar@biocon.com<br />

BMC Proceedings 2011, 5(Suppl 8):P95<br />

Introduction: The micro heterogeneity or quality of a protein has been<br />

shown to have a significant impact on its physical, chemical and biological<br />

properties both in vitro and in vivo [1]. Micro heterogeneity is evaluated in<br />

terms of post translational modifications such as glycosylation, charge<br />

variants, aggregates and fragments profile. The biggest challenge in<br />

Figure 2(abstract P94) The effect of different carbon sources on ECAR from lactic acid production and on metabolic shift in CHO_mAb. Change of<br />

baseline ECAR of CHO_mAb cells after Carbon/nitrogen source injection versus time (A). Glucose and mannose preference for aerobic glycolysis(B)<br />

Comparison of maximum ECAR in XF96 with lactic acid in shake flasks showed an excellent correlation, R 2 =0.91 (C).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P95) Plot of N.PCD vs. N.GL % for Ab1 & Ab3 suggest that there are some clones which have very different PCDs but similar product<br />

quality.<br />

process development is to find a balance between increasing productivity<br />

while maintaining product quality.<br />

In our study we focus on protein glycosylation, which is a process in which<br />

oligosaccharides are added to the protein during synthesis. There are<br />

multiple possible reactions in the pathway and it takes a long time for a<br />

glycosylated protein to be fully processed. If some protein molecules have a<br />

shorter residence time in the ER and Golgi, the glycan may be only at an<br />

intermediate stage [1]. For recombinant glycoprotein, increase in cell specific<br />

productivity (amount of product produced per cell per unit time) which may<br />

result in shorter residence time in the ER and Golgi, must be weighed<br />

against possible changes in product quality attributes like glycosylation [2].<br />

Ourstudyconcludesthatitispossible to produce a protein with desired<br />

product quality profile with high specific productivity. Two different clones<br />

with the same productivity can have different product quality profiles;<br />

alternatively, the same clone with different specific productivity can be<br />

manipulated to produce the same desired product quality by altering the<br />

cell culture parameters or addition of supplements. This observation also<br />

influences the acknowledged methodology for selecting clones with higher<br />

productivity while still maintaining their product quality profile. Various<br />

process manipulations were evaluated as an attempt to improve on the<br />

product quality profiles without compromising the productivity.<br />

Materials and methods: Three CHO cell lines (A, B & C) expressing three<br />

different Antibodies (Ab1, Ab2 & Ab3) were cultured in commercially<br />

available animal component free media in 125 ml Erlenmeyer shake flasks<br />

and BIOSTAT B-DCU lab bioreactors. Cell Count and Viability were analyzed<br />

by Cedex Hires (Innovatis) and heamocytometer using Trypan blue dye<br />

exclusion. The product concentration was determined by Affinity<br />

Figure 2(abstract P95) a: Profiles of Process 1, 2 & 3 for Ab1 Figure 2b: Profiles of Process A, B & C for Ab3.<br />

Page 128 of 181<br />

chromatography and characterization (Glycan profiling) by Normal phase<br />

HPLC.<br />

Results and discussion: Clone Selection program: Figure 1 shows the<br />

plot of N.PCD (normalized specific productivity – picogram per cell per<br />

day) vs. N.GL% (normalized values of one type of glycosylated species) of<br />

different clones for the antibodies Ab1 & Ab3. Both show a similar<br />

general trend indicating an increase in N. GL (%) with increasing specific<br />

productivities. There are however some exceptions where clones with<br />

significantly different specific productivity show very similar glycosylation<br />

profile, which suggest the role of process conditions in affecting the<br />

product quality.<br />

Case study 1: Ab1: As seen in (Figure 2a), the desired N. GL (%) for Ab1<br />

was comparable to the product obtained from the high PCD clones in<br />

Process 1. However, when the process was run in a different reactor<br />

configuration, a decrease in N.GL (%) was observed. Experiments were<br />

done to understand the impact of changes in the reactor conditions by<br />

varying the reactor dependent parameters (aeration, agitation etc) and<br />

the feeding strategy. These results were used to modify the Process 2<br />

andmadeasamorerobustProcess3.TheProcess3wasabletogivea<br />

higher value of N.GL (%) while still retaining the high PCD.<br />

Case study 2: Ab2: All the high producing clones for Ab2 were giving<br />

significantly higher N.GL (%) compared to the desired quality. A study<br />

was conducted to evaluate the possibility of choosing the high producing<br />

clone and manipulate the glycan profiles to be able to meet the product<br />

quality requirements. Intermittent samples were taken from the Fed<br />

batch runs and analyzed for product conc. and glycan profiles. Both PCD<br />

and N.GL (%) vary during the course of the run with a general trend of


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

higher N.GL(%) with increase in PCD. However there were exceptions like<br />

day 8 vs. day 12 where the PCD of day 8 was significantly lower than day<br />

12 however the N.GL(%) value was higherfortheday8.Thefeeding<br />

strategy and the process parameters (controlled and measured) around<br />

day 8 were estimated and compared to other days.<br />

The results of the investigation showed that a feed added at around day 8<br />

during the batch, helped in reducing the N. GL (%) values. The quantity of<br />

this feed as well as its feeding strategy were changed which helped in<br />

getting the desired product quality. The results from the study done above<br />

were implemented in the process.<br />

Case study 3: Ab3: The desired N. GL (%) for the product was significantly<br />

lower than the range obtained from most of the clones. The process used<br />

here is referred to as Process A. Feeding concepts implemented in the Ab2<br />

improved process were tested in the Ab3 which is referred as Process B.<br />

The feed manipulations helped in bringing down the N.GL (%) values.<br />

However they were still much higher than the desired range. The time<br />

course analysis of the Process B batches was done. The analysis indicated<br />

some days during the run where the level of N.GL (%) were much lower<br />

compared to other days. Detailed analysis of the culture conditions during<br />

these days was done. The outcome of these analysis were implemented in<br />

the process by changing the culture parameters during the run. This<br />

process is referred as Process C. The profiles of batches run with Processes<br />

A, B and C are shown in (Figure 2b). Significant decrease in the level of N.<br />

GL (%) was observed in Process C without affecting the PCD of the cells.<br />

Conclusion: It was generally observed that the clones with high specific<br />

productivity also have high levels of N.GL (%). Since some of the<br />

products required lower levels of N.GL (%); these high producing clones<br />

Page 129 of 181<br />

were found to be unsuitable. However, the results of this study show that<br />

with modified conditions of process parameters and feeding strategies, it<br />

was possible to alter the N.GL (%) levels without impacting the cell line<br />

specific productivity. These factors when taken into consideration during<br />

clone selection may still allow the selection of high producing clones<br />

with a possibility to alter the product profiles during development.<br />

Acknowledgement: Cell Culture group: Chandrashekhar Kuruvangi,<br />

Janani Kanakrajan, Rohit Diwakar<br />

References<br />

1. Hu WS, Betenbough M: Technology for Secretory Therapeutic Proteins.<br />

Cellular Bioprocess technology 2007.<br />

2. Hossler P, Khattak SF, Li Zheng: Optimal and consistent protein<br />

glycosylation in mammalian cell culture. Glycobiology 2009, 19(9):936-949.<br />

P96<br />

A novel peptide to enhance recombinant BMP-2 production in<br />

mammalian cell cultures<br />

Aileen J Zhou 1* , Cameron ML Clokie 1,2 , Sean AF Peel 1,2<br />

1 Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University<br />

of Toronto, Toronto, Ontario Canada, M5G 1G6; 2 Induce Biologics Inc,<br />

Toronto, Ontario, Canada, M5R 3N8<br />

E-mail: aileen.zhou@dentistry.utoronto.ca<br />

BMC Proceedings 2011, 5(Suppl 8):P96<br />

Background: Due to their osteoinductive properties, recombinant human<br />

bone morphogenetic proteins (rhBMPs) have been used successfully for<br />

Figure 1(abstract P96) After 24 h incubation with or without IND-1, the amount of proBMP-2 and mature BMP-2 proteins in (A) CHO and (B) HEK<br />

conditioned media were measured by proBMP-2 and BMP-2 ELISA. Data are presented as mean ± SEM (n = 3 experiments) (**P


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

bone regeneration and replacement. However, the yields rhBMPs yields in<br />

mammalian expression systems are very low, resulting in their high cost.<br />

BMPs are synthesized as a precursor, proBMP, which undergoes enzymatic<br />

cleavage by proprotein convertases (PCs) to form the mature BMP [1]. Furin,<br />

an enzyme of the PC family, has shown to cleave BMP-4 [2] and BMP-2<br />

(Zhou et al., unpublished data). This study investigated the effect and<br />

mechanism of action of polyarginine furin inhibitor, IND-1, on rhBMP-2<br />

production in mammalian cell lines overexpressing rhBMP-2.<br />

Materials and methods: Two stable cell lines expressing the hBMP2 gene,<br />

CHO-BMP2 and HEK-BMP2, were cultured in the presence of IND-1 in shortterm<br />

(24 h, multi-well) and long-term (two-month, perfusion flasks)<br />

cultures. The rhBMP-2 produced was characterized by Western blot and its<br />

activity assessed using the C2C12 cell-based assay. The amount of proBMP-<br />

2 and mature BMP-2 produced was quantified by ELISA. The mRNA level of<br />

BMP-2 and furin in cells treated with or without IND-1 was compared by<br />

real-time RT-PCR. Cellular uptake of IND-1 was estimated by measuring the<br />

fluorescence of cell lysates following incubation with FITC labeled IND-1.<br />

Cellular PC activity post IND-1 incubation was measured using the Boc-<br />

RVRR-AMC substrate. Furin-specific siRNA was used to knock down the<br />

furin expression in CHO-BMP2 cells and its effect on the rhBMP-2<br />

production was determined.<br />

Results: Stably transfected CHO-BMP2 cells secreted 36 kDa rhBMP-2<br />

dimers that were biologically active. In 24 h cell cultures, IND-1 treated<br />

cells produced significantly greater amounts of proBMP-2 (≥ 10-fold,<br />

P


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P97) Viable cell density (VCD) and cell viabilities of CHO cells cultured in a fed-batch bioreactor<br />

process<br />

Process time (d) VCD mean (cells/mL) STDV CV (%) Viability mean (%) STDV CV (%)<br />

Cedex 0.8 7.33E+05 1.35E+04 1.8 96.39 0.92 1.0<br />

3.8 4.18E+06 5.94E+04 1.4 97.01 0.33 0.3<br />

MACSQuant Analyzer 0.8 6.47E+05 9.42E+03 1.5 95.30 0.03 0.0<br />

3.8 4.41E+06 2.87E+04 0.7 96.06 0.06 0.1<br />

Vi-Cell 0.8 7.11E+05 1.06E+05 14.9 96.31 0.73 0.8<br />

3.8 5.03E+06 9.92E+04 2.0 96.98 0.06 0.1<br />

Figure 1(abstract P97) (A) The time course of an unstable MTX amplification is presented. A decrease in the fraction of IgG-producing cells (F P) is caused<br />

by resistance to increasing MTX concentrations (c MTX). (B) Dot plots for the samples at 307 h (160 nM MTX) and 687 h (640 nM) are shown. IgG-producing<br />

cells appear in the upper right gates.<br />

The MACSQuant Analyzer allows the determination of cell density and<br />

viability with high reproducibility. The mean data were comparable to<br />

common cell-counting systems. The results obtained with the MACSQuant<br />

Analyzer, however, showed better accuracy as reflected by the lower CV.<br />

Moreover, through its FSC, SSC, and seven fluorescence channels, as well as<br />

its three lasers it allows sophisticated, multiparametric flow cytometric<br />

analysis.<br />

References<br />

1. Cacciatore , Chasin , Leonard : Gene amplification and vector engineering to<br />

achieve rapid and high-level therapeutic protein production using the<br />

Dhfr-based CHO cell selection system. Biotechnol Adv 2010, 28(6):673-681.<br />

2. Jun , Kim , Baik , Hwang , Lee : Selection strategies for the establishment of<br />

recombinant Chinese hamster ovary cell line with dihydrofolate reductasemediated<br />

gene amplification. Appl Microbiol Biotechnol 2005, 69(2):162-169.<br />

P98<br />

Bioreactor cultivation of CHO DP-12 cells under sodium butyrate<br />

treatment – comparative transcriptome analysis with CHO cDNA<br />

microarrays<br />

Sandra Klausing *† , Oliver Krämer † , Thomas Noll<br />

Institute of Cell Culture Technology, Bielefeld University, 33615 Bielefeld,<br />

Germany<br />

E-mail: skl@zellkult.techfak.uni-bielefeld.de<br />

BMC Proceedings 2011, 5(Suppl 8):P98<br />

Page 131 of 181<br />

Background: Sodium butyrate (NaBu) is not only known to inhibit<br />

proliferation but also to increase the specific productivity in cultivation of<br />

Chinese hamster ovary (CHO) cells [1] – the most commonly used<br />

mammalian cell line for pharmaceutical protein production [2]. So far,<br />

little is known about the underlying mechanisms and genes that are<br />

affected by butyrate treatment. Besides the proteomic approach to<br />

unravel proteins involved in the processes, the analysis of transcriptomes<br />

presents another promising method. Here we show an application of our<br />

CHO cDNA microarray to identify genes associated with increased<br />

productivity during cultivation of CHO cells under sodium butyrate<br />

treatment.<br />

Materials and methods: Four batch cultivations of CHO DP-12 cells<br />

(clone # 1934, ATCC CRL-12445) were performed in 2 L bioreactor systems<br />

under pO 2- and pH-controlled conditions. In the exponential growth<br />

phase, 67 hours after inoculation, 2 mM sodium butyrate was added to<br />

three processes. The fourth was left untreated to function as control<br />

culture. Samples were taken before and then repeatedly after the addition<br />

of butyrate. RNA was isolated from cell pellets of 5·10 6 cells using TRIzol®<br />

Reagent (Invitrogen). For subsequent cDNA labeling, the Agilent Low-Input<br />

QuickAmp Labeling Kit (Agilent Technologies) was used. The custom<br />

designed 2 x 105 k cDNA microarray (Agilent Technologies) was spotted<br />

with 94,580 probes designed from CHO cDNA sequenced in-house. 38,310<br />

of 41,039 sequenced contigs were used for the microarray, each covered<br />

by 2-4 probes [3]. Data analysis was done with ArrayLims, EMMA, and<br />

SAMS, three CeBiTec based software tools [4]. The raw data gathered by


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P98) (A): Concentration of viable cells and cell viabilities for the time course of CHO DP-12 batch processes. Error bars represent the<br />

standard deviation of triplicate measurements with the Cedex system (Roche Diagnostics). Red and orange lines represent biological replicates of cultures<br />

treated with 2 mM NaBu, the control process is shown in green. Dashed lines show viabilities. The grey arrow indicates the addition of NaBu, the grey<br />

circles show the sample points compared later in the microarray analysis (72 h of NaBu Treatment). (B): Number of up- and downregulated genes of<br />

selected KEGG pathway categories with a detailed view of four pathways from the Cell Growth and Death KEGG category. Results show only those found<br />

as differentially expressed after filtering. Red: downregulated in NaBu cultures; green: upregulated in NaBu cultures (compared to control culture).<br />

the microarray experiments were processed by standard Agilent<br />

background normalization and subsequent lowess normalization.<br />

Results: The control culture reached a maximum viable cell density of<br />

1·10 7 cells/mL while NaBu treated cells reached a plateau at about 6·10 6<br />

cells/mL and retained a viability above 90% four days longer than<br />

untreated cells (Figure 1A). The three biological replicates of NaBu cultures<br />

yielded results with similar general trends. The maximum antibody<br />

concentration of the control culture was 110 mg/L whereas cells treated<br />

with NaBu reached a maximum of 175 mg/L antibody. 72 hours after<br />

addition of NaBu the specific antibody production rate was increased by<br />

a factor of 3.6 (NaBu culture: 4.5 pg/(cell·d)) compared to control culture<br />

(1.2 pg/(cell·d)).<br />

Of this time point, samples were analyzed in microarray experiments. A<br />

significance test with FDR control (a=0.05) was carried out for the four<br />

technical replicates (including two dye-swaps) of the microarray. For<br />

analysis, the following filtering settings were chosen to identify differentially<br />

expressed genes: adjusted p-value ≤ 0.05, log-ratio < -1 or > 1 (equals fold<br />

change < -2 or > 2) and log-intensity ≥ 6 (equals ≥ 64 raw intensity). From a<br />

total of 1461 genes found to be differentially expressed under NaBu<br />

treatment, 771 genes were upregulated and 690 genes were downregulated<br />

Table 1(abstract P98) Fold change of selected genes from microarray analysis<br />

(derived from EC numbers in KEGG pathways, Figure 1B). Many differentially<br />

expressed genes from pathways involved in carbohydrate, lipid, amino acid<br />

and glycan metabolism are upregulated which is most likely linked to higher<br />

productivity. A large portion of genes from pathways associated with cell<br />

growth and death are downregulated and most of these genes originate<br />

from cell cycle processes. This correlates with reports of cell cycle arrest<br />

under NaBu treatment [1]. Some examples of regulated genes are shown in<br />

Table 1.<br />

Conclusions: Microarray analysis revealed a high number of regulated<br />

genes under sodium butyrate treatment in pathways like carbohydrate<br />

metabolism, cell cycle and signal transduction. Some of the regulated<br />

genes are promising targets for overexpression or knockdown/knockout<br />

experiments and we will further investigate the knockdown effect of<br />

selected genes using a siRNA approach in CHO cells. Our in-house<br />

microarray is suitable for further transcriptomic analysis of CHO cells<br />

under various conditions.<br />

Acknowledgements: We would like to thank E. Schulte-Berndt (Institute<br />

for Genome Research and Systems Biology, CeBiTec, Bielefeld) for help<br />

with microarray hybridization and O. Rupp (Bioinformatics Resource<br />

Facility, CeBiTec, Bielefeld) for help with microarray data analysis.<br />

KEGG pathway category Gene symbol Description Mean fold change<br />

in NaBu culture<br />

compared to<br />

control culture<br />

Page 132 of 181<br />

# of probes<br />

CA150 Transcription factor CA150b -3.31 ↓ 3<br />

Transcription & Translation Ccdc12 Coiled-coil domain containing 12 -2.01 ↓ 1<br />

Y14 RNA-binding protein 8A 2.14 ↑ 2<br />

Pkmyt1 Protein kinase, membrane associated tyrosine/threonine 1 -2.04 ↓ 2<br />

Cell Cycle c-Myc Myc proto-oncogene protein -3.44 ↓ 3<br />

Ink4c Cdkn2c cyclin-dependent kinase inhibitor 2C (p18, inhibits CDK4) 3.04 ↑ 2<br />

CycD Cyclin D1 (Ccnd1) 2.49 ↑ 1<br />

Pdcd4 Programmed cell death 4 3.89 ↑ 2<br />

Apoptosis Casp6 Caspase 6 3.06 ↑ 3<br />

PI3K Phosphatidylinositol 3-kinase, regulatory subunit, polypeptide 1 2.44 ↑ 2


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

References<br />

1. Kumar N, Gammell P, Clynes M: Proliferation control strategies to improve<br />

productivity and survival during CHO based production culture: A<br />

summary of recent methods employed and the effects of proliferation<br />

control in product secreting CHO cell lines. Cytotechnology 2007, 53(1-<br />

3):33-46.<br />

2. Jayapal K, Wlaschin K, Yap M, Hu W-S: Recombinant protein therapeutics<br />

from CHO cells - 20 years and counting. Chem. Eng. Prog. 2007,<br />

103(10):40-47.<br />

3. Becker J, Hackl M, Jakobi T, Rupp O, Timmermann C, Szczepanowski R,<br />

Borth N, Goesmann A, Grillari J, Noll T, Pühler A, Tauch A, Brinkrolf K: Next-<br />

Generation Sequencing of the CHO cell transcriptome. BMC Proceedings<br />

2011, 5(Suppl 8):P6.<br />

4. Center for Biotechnology, IFB - Institute for Bioinformatics: Computational<br />

Genomics, Software. [http://www.cebitec.uni-bielefeld.de/cebitec/<br />

computational-genomics/software.html].<br />

P99<br />

Characterization of chromatographic yeast extract fractions promoting<br />

CHO cell growth<br />

Mathilde Mosser 1,2 , Romain Kapel 1 , Arnaud Aymes 1 , Laurent-Michel Bonanno 2 ,<br />

Eric Olmos 1 , Iris Besançon 2 , Dominique Druaux 2 , Isabelle Chevalot 1 , Ivan Marc 1 ,<br />

Annie Marc 1*<br />

1 Laboratoire Réactions et Génie des Procédés, UPR CNRS 3349, Nancy-<br />

Université, Vandœuvre-lès-Nancy, France; 2 Bio Springer, Maisons-Alfort,<br />

France<br />

E-mail: annie.marc@ensic.inpl-nancy.fr<br />

BMC Proceedings 2011, 5(Suppl 8):P99<br />

Background: Many studies underline the great benefits of yeast extracts<br />

(YE), used as supplements in animal free culture media, on cell growth and<br />

recombinant protein production [1,2]. Nevertheless, their unknown<br />

composition and batch-to-batch variability of commercial YE remain<br />

constraints for industrial processes [3,4]. Consequently, the identification of<br />

bioactive YE molecules is challenging for process reliability. The main<br />

strategy to respond upon this problem is to fractionate the extract and then<br />

to characterize the fractions. So far, several fractionation processes such as<br />

ultrafiltration [5], gel filtration chromatography [6] or alcoholic precipitation<br />

[7] have been investigated. These studies suggested that the active<br />

components were mainly small molecules (< 1000 Da). But, the other<br />

physico-chemical properties of YE components have been poorly studied<br />

until now. In regards of this statement, it is proposed to get better<br />

knowledge of the YE molecules leading to an improvement of CHO cell<br />

growth, by implementing various chromatographic fractionation processes.<br />

Material and methods: CHO-AMW cell line producing anti-human RhD-<br />

IgG1 was provided by Dr. F. Wurm. Cell growth assays were performed in<br />

96-well plates with 200 μL of working volume inside an incubator at 37 °C<br />

and 5% CO 2. Cells were cultivated in a protein-free chemically defined<br />

medium RPMI 1640/BDM (90/10) supplemented with 4 mM of glutamine<br />

(Eurobio, France) and supplemented or not by 1 g.L -1 of YE fraction. Cell<br />

concentration was in-situ monitored by Cellscreen® analyser (Innovatis,<br />

Germany).<br />

Raw YE, the soluble fraction of yeast autolysate, was provided by<br />

BioSpringer. A nanofiltration step using Nanomax 50 membrane (Millipore,<br />

Bedford, MA) was used to remove the very small molecules such as salts<br />

and amino acids. YE was first fractionated by three one-step preparative<br />

chromatographies according to the described conditions (Table 1).<br />

Fractions containing salts and buffers were purified and concentrated by<br />

nanofiltration. In another way, a complete chromatographic fractionation<br />

was achieved by sequentially separating YE with anion-exchange,<br />

hydrophobic interaction and size exclusion chromatography. All fractions<br />

were lyophilized for further analyses and cell culture assays.<br />

Concentration of total amino acids, polysaccharides and nucleic acids<br />

were determined in all fractions by HPLC methods after total hydrolysis.<br />

Molecular weights were evaluated by size-exclusion HPLC [8].<br />

Results: Firstly, the YE was separately fractionated either by anion<br />

exchange (AEC), hydrophobic interaction (HIC) or gel filtration<br />

chromatography (GFC). The AEC allowed isolating in one fraction the<br />

nucleic acids strongly bound to the gel, while peptides were distributed<br />

among all fractions depending on their electrical charges. In the HIC<br />

fractions, important quantities of buffer salts still remained despite the<br />

desalting process. Finally, the GFC led to three fractions containing various<br />

proportions of nucleic acids, polysaccharides and peptides. Then, the<br />

influence of the fractions on the CHO cell growth was evaluated in 96-well<br />

microplates (Figure 1-A). All GFC fractions presented similar results than<br />

raw YE. The HIC fractions did not stimulate the cell growth, probably due<br />

to residual buffer salts. Furthermore, only one fraction from AEC, devoid of<br />

nucleic acids but enriched in positive-charged peptides, presented a<br />

similar impact than YE.<br />

Secondly, YE was divided in 18 fractions by a global process including the<br />

three chromatographic methods in a sequential mode: AEC, HIC, GFC.<br />

The final GF step presented the advantage to remove the residual salts.<br />

The cell growth monitoring pointed out two fractions, which led to similar<br />

results than the YE (Figure 1-B). These fractions contained peptides, mainly<br />

positively charged and polar, low quantities of polysaccharides but no<br />

nucleic acids. The amino acids quantification of fraction A.1-H.1-W.3<br />

underlined that peptides were mainly composed of lysine and arginine.<br />

This result was consistent with a recent patent claiming that C-terminal<br />

arginine containing tripeptides were at least partially responsible for the<br />

growth promoting activity of a peptone (9).<br />

Conclusion: A one step-fractionation process by anion exchange<br />

chromatography led to a simplified composition of raw YE without<br />

compromising its stimulating effect on CHO cell growth. Furthermore, the<br />

three-step fractionation process allowed a better knowledge of the physicochemical<br />

properties of the molecules involved in cell growth stimulating<br />

effects: polar positively charged and mainly composed of peptides enriched<br />

in arginine and lysine residues.<br />

References<br />

1. Sung YH, Lim SW, Chung JY, Lee GM: Yeast hydrolysate as a low-cost<br />

additive to serum-free medium for the production of human<br />

thrombopoietin in suspension cultures of Chinese hamster ovary cells.<br />

Appl Microbiol Biot 2004, 63:527-536.<br />

2. Kim DY, Lee JC, Chang HN, Oh DJ: Development of serum-free media for<br />

a recombinant CHO cell line producing recombinant antibody. Enzyme<br />

Microb Tech 2006, 39:426-433.<br />

3. Butler M: Animal cell cultures: recent achievements and perspectives in<br />

the production of biopharmaceuticals. Appl Microbiol Biot 2005,<br />

68:283-291.<br />

4. Grillberger L, Kreil TR, Nasr S, Reiter M: Emerging trends in plasma-free<br />

manufacturing of recombinant protein therapeutics expressed in<br />

mammalian cells. Biotechnol J 2009, 4:186-201.<br />

5. Chun BH, Kim JH, Lee HJ, Chung N: Usability of size-excluded fractions of<br />

soy protein hydrolysates for growth and viability of Chinese hamster<br />

ovary cells in protein-free suspension culture. Bioresource Technol 2007,<br />

98:1000-1005.<br />

6. Mendonça RZ, Oliveira EC, Pereira CA, Lebrun I: Effect of bioactive<br />

peptides isolated from yeastolate, lactalbumin and NZCase in the insect<br />

cell growth. Bioprocess Biosyst Eng 2007, 30:157-164.<br />

7. Shen CF, Kiyota T, Jardin B, Konishi Y, Kamen A: Characterization of<br />

yeastolate fractions that promote insect cell growth and recombinant<br />

protein production. Cytotechnology 2007, 54:25-34.<br />

Table 1(abstract P99) Operating conditions of preparative chromatographic fractionations<br />

Separation Gel / Column Fractions<br />

Anion-exchange Q Sepharose, H 10 cm, j 10 cm A.1, A.2, A.3<br />

Hydrophobic interaction Phenyl Sepharose , H 10 cm, j 5 cm H.1, H.2<br />

Size-exclusion G-15 Sephadex, H 75 cm H, j 5 cm W.1, W.2, W.3<br />

Sequentially three step process same than one step processes A.x-H.y-W.z<br />

Page 133 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P99) Maximal concentration of CHO cells with or without 1 g.L -1 of YE fractions obtained (A) by one-step and (B) three-step<br />

chromatography. The control culture was performed in the chemically-defined medium.<br />

8. Chabanon G, Alves da Costa L, Farges B, Harscoat C, Chenu S, Goergen JL,<br />

Marc A, Marc I, Chevalot I: Influence of the rapeseed protein hydrolysis<br />

process on CHO cell growth. Bioresource Technol 2008, 99:7143-7151.<br />

9. Wilkins J: Biologically active c-terminal arginine-containing peptides.<br />

United states patent application 2009, US 2009/0143248 A1.<br />

P100<br />

Study of expansion of porcine bone marrow mesenchymal stem cells<br />

on microcarriers using various operating conditions<br />

Caroline Ferrari 1 , Frédérique Balandras 1 , Emmanuel Guedon 1 , Eric Olmos 1 ,<br />

Nguyen Tran 2 , Isabelle Chevalot 1 , Annie Marc 1*<br />

1 Laboratoire Réactions et Génie des Procédés, UPR-CNRS 3349, Nancy-<br />

Université, Vandœuvre-lès-Nancy, France; 2 École de Chirurgie, Faculté de<br />

Médecine, Vandœuvre-lès-Nancy, France<br />

E-mail: annie.marc@ensic.inpl-nancy.fr<br />

BMC Proceedings 2011, 5(Suppl 8):P100<br />

Background: Bone marrow mesenchymal stem cells (BM-MSCs) represent<br />

promising source for tissue engineering and cell therapy, due to their<br />

multipotency, immunoregulation and self-renewal properties [1,2]. The<br />

expansion phase of these cells prior to differentiation and/or injection to the<br />

patient remains a critical step. BM-MSCs are classically expanded in small<br />

scale culture systems, with low control of culture conditions. However, tissue<br />

engineering and cell therapy require very large quantities of cells that<br />

cannot be easily achieved using processes in static flasks [3]. Microcarriers,<br />

classically used for industrial large scale culture of continuous cell lines,<br />

could be advantageously applied to stem cells expansion such as BM-MSCs<br />

[4-6]. Our objective was to study the influence of some operating<br />

parameters (agitation rate, microcarrier feed) on expansion and organization<br />

(adhesion, aggregation) of porcine BM-MSCs cultivated on collagen-free<br />

microcarriers, in order to improve cell expansion while maintaining their<br />

multipotency.<br />

Materials and methods: BM-MSCs were extracted from the iliac crest of<br />

bone marrow of three month old pigs, and purified by adherence and selfrenewal<br />

properties. Cells were expanded in T-flask and on 1.2 g/L Cytodex<br />

1 carriers (GEHealthcare) in spinner flasks. Initial cell density was 6000 cell/<br />

cm 2 and 30 000 cell/mL (total surface of 1000 cm 2 and medium volume of<br />

200 mL). Culture medium was a modified a-minimal essential medium (a-<br />

MEM, Sigma) supplemented with 10% fetal bovine serum (FBS). All cultures<br />

were performed inside an incubator (37 °C; 5% CO2). Medium was<br />

exchanged (50% volume) every two days starting from day three. Cell<br />

nuclei were counted by flow cytometry (Guava Easycyte) after cell lysis by<br />

citric acid. Cell adhesion and aggregation were observed by optical<br />

microscopy (x 40) after methylene blue staining. Cell multipotency was<br />

assayed by using differentiation kits (Invitrogen).<br />

Results: Prior to study the cell expansion phase, the adequate operating<br />

conditions of the seeding phase were determined such as the cell to bead<br />

ratio and the medium composition. Then, kinetics of cell expansion was<br />

compared between T-flask and spinner flask with cells on mirocarriers.<br />

Page 134 of 181<br />

After 8 days, BM-MSCs reached the same maximal total cell nuclei in both<br />

culture systems until cells remained as monolayer in T-flask but<br />

aggregated on microcarriers. As a consequence, cell density seemed to<br />

decrease on microccariers due to cell aggregation (Figure 1).<br />

In a second part, growth kinetic studies of porcine BM-MSCs attached on<br />

Cytodex 1 carriers were performed at different agitation rates (0, 25, 75<br />

rpm) in spinner flasks. Under stirred conditions, BM-MSCs cell population<br />

reached a maximal cell concentration (1.5 x 10 5 cell/mL; x 5 multiplication<br />

factor) before to decline whatever the agitation rate used. Small<br />

aggregates was observed on microcarrier surface in the early stage of the<br />

cultures. Once those aggregates reached a critical size, they left from the<br />

microcarriers and remained in suspension, where no more growth could<br />

be observed. However, culture without agitation reached a similar maximal<br />

cell density but a longer steady phase was observed during 300 hours until<br />

cell/microcarrier clusters occurred at day 13. As cell/cell aggregation was<br />

notobservedat0rpm,itcouldbeassumed that Cytodex 1 surface was<br />

not directly involved in the cell aggregation phenomenon, which seemed<br />

promoted under agitation.<br />

To verify if the aggregates were able to dissociate when exposed to new<br />

surfaces, cells were cultivated on agarose in static mode to favor<br />

aggregates. Those aggregates dissociated once exposed to static surfaces<br />

such as T-flasks or fresh Cytodex 1 carriers. Based on these observations<br />

and to allow homogeneous and controlled stirred cultures, the addition<br />

of fresh carriers during stirred cultures was further evaluated.<br />

Firstly, fresh microcarriers were added in stirred spinner flask at day 10,<br />

when cell aggregates were already formed and cell density decreasing.<br />

Cells were able to colonize the new added surface, and to grow again,<br />

showing that cell aggregation can be reversed by adding fresh carriers. In<br />

a second time, fresh carriers were sequentially added at day 4, 8 and 12.<br />

As a result, after 12 days, total cell nuclei reached values 3 times higher<br />

than in the culture without carrier addition, suggesting that cell<br />

aggregation could be prevented by early addition of fresh carrier in the<br />

stirred culture.<br />

Following expansion in T-flask or on microcarriers, BM-MSCs were<br />

harvested by trypsination and tested for their multipotency. Their similar<br />

ability to differentiate in adipocytes, chondrocytes and osteocytes<br />

indicated that the multipotency was preserved after cell expansion on<br />

Cytodex 1 carriers.<br />

Conclusion: BM-MSCs culture on Cytodex-1 collagen-free carriers in<br />

stirred systems allowed the cell expansion while maintaining their<br />

multipotency. By adding fresh microcarriers, cell aggregation could be<br />

prevented and the expansion phase duration extended. This culture<br />

process is expected to be transferred to a larger controlled culture<br />

system, in order to fulfill the need of high quantities of multipotent<br />

BM-MSCs.<br />

References<br />

1. Caplan A: Why are MSCs therapeutic? New data: new insight. J Pathol<br />

2009, 217:318-324.<br />

2. Chamberlain G, Fox J, Ashton B, Middleton J: Concise review:<br />

mesenchymal stem cells: their phenotype, differentiation capacity,


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P100) Porcine BM-MSCs growth on 1000 cm 2 curface of Cytodex 1 stirred at 25 rpm. Addition of fresh carriers at day 10 ; 50 %<br />

medium change every 2 days.<br />

immunological features, and potential for homing. Stem Cells 2007,<br />

25:2739-2749.<br />

3. Godora P, McFarland C, Nordon R: Design of bioreactors for mesenchymal<br />

stem cell tissue engineering. J Chem Technol Biotechnol 2008, 83:408-420.<br />

4. Frauenschuh S, Reichmann E, Ibold Y, Goetz P, Sittinger M, Ringe J: A<br />

microcarrier-based cultivation system for expansion of primary<br />

mesenchymal stem cells. Biotechnol Prog 2007, 23:187-193.<br />

5. Schop D, Janssen F, Borgart E, de Bruijn J, van Dijkhuizen-Radersma R:<br />

Expansion of mesenchymal stem cells using a microcarrier-based<br />

cultivation system: growth and metabolism. J Tissue Eng Regen Med 2008,<br />

2:126-135.<br />

6. Sart S, Schneider Y-J, Agathos S: Ear mesenchymal stem cells: an efficient<br />

adult multipotent cell population fit for rapid and scalable expansion.<br />

J Biotechnol 2009, 139:291-299.<br />

P101<br />

Growth and death kinetics of CHO cells cultivated in continuous<br />

bioreactor at various agitation rates<br />

Frédérique Balandras, Eric Olmos * , Caroline Hecklau, Fabrice Blanchard,<br />

Emmanuel Guedon, Annie Marc<br />

Laboratoire Réactions et Génie des Procédés, UPR CNRS 3349, Nancy-<br />

Université, Vandœuvre-lès-Nancy, France<br />

E-mail: eric.olmos@ensic.inpl-nancy.fr<br />

BMC Proceedings 2011, 5(Suppl 8):P101<br />

Background: Mammalian cells are known to be sensitive to hydrodynamic<br />

stresses. Therefore, soft agitation and aeration are generally recommended<br />

in culture bioreactor to prevent cell damages. Nevertheless, at the industrial<br />

scale, this may induce CO 2 accumulation, high mixing times, poor air<br />

dispersion and concentration gradients. In batch reactor, we have previously<br />

found that growth kinetics of CHO cells could be enhanced by increasing<br />

stirring frequencies from 80 to 600 rpm. However, these studies were<br />

limited in time due to depletion of substrates leading to a cell death which<br />

is not associated to hydrodynamic constraints. To overcome the effects on<br />

cell death from those nutritional limitations, in this work, longer stationary<br />

Page 135 of 181<br />

cultures of CHO cells were carried out in continuous mode in a 2-liter<br />

reactor operating with various stirring profiles.<br />

Materials and methods: CHO 320 cells were grown in a serum-free<br />

medium PF-BDM in a sparged and stirred tank reactor (1.4 L working<br />

volume; pO2: 50%; pH: 7.4; temperature: 37°C). Continuous culture was<br />

started in mid-exponential phase (D=0.02 h -1 ) and two agitation rate profiles<br />

were applied : 300-600-300-600 rpm and 600-300-600-800-900-1000 rpm.<br />

Viable and necrotic cell densities were estimated according to the Trypan<br />

blue exclusion method. Lysed cells were measured via the LDH release<br />

assay, whereas apoptotic cells were analysed by using Guava Easycyte<br />

cytometer. Glucose, lactate and glutamine concentrations were assayed with<br />

enzymatic commercial kits (Ellitech, Biomerieux, R-Biopharma) and ammonia<br />

concentration with a selective probe (Orion). Bioreactor hydrodynamics was<br />

numerically simulated by using Computational Fluid Dynamics to calculate<br />

the power dissipation rates and the velocity fields at each agitation rate<br />

(Fluent 6.3). The experimental validations were performed by Laser Doppler<br />

Velocimetry.<br />

Results:<br />

Continuous culture performed with the first agitation rate profile :<br />

300-600-300-600 rpm: Based on previous batch culture results [1], the<br />

batch phase was started at 300 rpm during 48 h in order to obtain the<br />

mid-exponential cell growth phase before starting the continuous mode<br />

(Figure 1A). Viable cell concentration increased to 30.10 5 cells.mL -1 at the<br />

steady state, while dead and lysed cells reached an average level of 5.10 5<br />

cells.mL -1 . A step of agitation rate from 300 to 600 rpm induced a 70%<br />

decrease in viable cells and a doubled dead cell concentration. The same<br />

variation was obtained for the second step of the experiment (300 rpm to<br />

600 rpm) but with higher dead and lysed cell concentrations of 13.10 5<br />

cells.mL -1 . Kinetics of glucose and glutamine consumption were monitored<br />

during the culture (Figure 1B). After 100 h of continuous culture, glutamine<br />

concentration decreased down to a limiting level around 0.03 g.L -1 and<br />

remained constant all over the experiment. Following the step from 300 to<br />

600 rpm, glucose concentration increased from 0.1 to 0.6 g.L -1 along with<br />

the viable cell concentration decrease from 28.10 5 to 8.10 5 cells.mL -1 .<br />

Consequently, no glucose limitation occurred at 600 rpm. Under<br />

hydrodynamic stresses, cell death mayoccureitherbynecrosisor<br />

apoptosis mechanisms [2,3]. Necrosis is a sudden phenomenon followed


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P101) Kinetics of continuous cultures performed with two different profiles of agitation rates (—): (A, C) viable cells (-⃟-), dead cells<br />

(-⃞-), lysed cells (-Δ); (B, D) glucose (-⃞-) and glutamine (-Δ-).<br />

by a rapid cell lysis, whereas apoptosis involves enzymatic cascades<br />

reactions leading to molecular, enzymatic and structural properties<br />

modifications. In our study, analysis of dead cell populations by using flow<br />

cytometry revealed that cells mostly died by apoptosis for the first step<br />

from 300 to 600 rpm, while necrosis became the predominant death<br />

mechanism after the second step (Table 1).<br />

Continuous culture performed with the second agitation rate profile<br />

: 600-300-600-800-900-1000 rpm: For the second run, the batch phase<br />

was started at 600 rpm since such an agitation rate was shown not to<br />

be deleterious to CHO cells cultivated in batch mode [1]. In this case,<br />

the cells reached an average concentration of 30.10 5 cells mL -1 at the<br />

steady state of the continuous culture (Figure 1C). However, lysed cells<br />

accumulatedupto8.10 5 cells.mL -1 after 200 h of culture. The first<br />

change from 600 to 300 rpm did not significantly influence the viable<br />

cell density, while a slight decrease of lysed cell concentration was<br />

observed. It has to be noted that the increase in the stirring rate from<br />

600 to 1000 rpm did not lead to massive cell death. While the viable<br />

Page 136 of 181<br />

cell density decreased and remained at 20.10 5 cells.mL -1 ,thelysed<br />

cell concentration gradually increased to reach 10.10 5 cells.mL -1 and<br />

the dead cell density (Trypan blue) remained negligible at 2.10 5 cells.<br />

mL -1 . In these conditions, no glucose and glutamine limitation was<br />

observed (Figure 1D). Dead cell analysis by Guava cytometry indicated a<br />

maximal apoptotic cell level around 5 to 10% of total cells throughout<br />

the culture (Table 1). Necrotic cells appeared only at 800 rpm and<br />

accounted to 40% of the total cells at 1000 rpm. Then, cell death<br />

occurred essentially by lysis and necrosis during this second continuous<br />

culture.<br />

Discussion: When CHO cells were cultivated in a continuous mode and<br />

subjected to an increase in power dissipation rates from 0.32 (300 rpm)<br />

to 2.5 W.kg -1 (600 rpm), massive cell necrosis and apoptosis occurred,<br />

while this cellular response was not observed in batch mode for the<br />

same levels of agitation. Therefore, the CHO cell death could be<br />

attributed to the observed glutamine limitation known to induce<br />

apoptosis [4]. Furthermore, cells initially cultivated with a higher


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P101) Percentages of apoptotic and necrotic cells measured by using a GUAVA cytometer<br />

Time (h) RPM Necrotic cells (%) Apoptotic cells (%)<br />

Culture 1 Culture 2 Culture 1 Culture 2 Culture 1 Culture 2 Culture 1 Culture 2<br />

50 50 300 600 7 4 2 6<br />

100 150 300 300 19 2 2 4<br />

150 340 600 300 9 1 15 4<br />

180 480 600 600 2 2 31 7<br />

280 600 300 600 4 2 34 8<br />

320 675 300 800 23 8 8 5<br />

380 745 300 800 21 10 5 8<br />

430 825 600 900 18 30 6 10<br />

450 865 600 1000 49 40 16 13<br />

dissipation rate (600 rpm, 2.5 W.kg -1 ) revealed a better ability to resist to<br />

very high dissipation rates (1000 rpm, 11 W.kg -1 ), suggesting cell<br />

adaptation or selection. In conclusion, the growth and death<br />

mechanisms of CHO cells cultivated at high dissipation rates strongly<br />

depend on the sequence of power dissipation levels and on the<br />

availability of nutrients. Chemostat could be thus an appropriate system<br />

to better understand the complex cellular response to hydrodynamic<br />

stresses.<br />

References<br />

1. Barbouche N: Réponse biologique de cellules animales à des contraintes<br />

hydrodynamiques: simulation numérique, expérimentation et<br />

modélisation en bioréacteurs de laboratoire. PhD thesis, Nancy-University,<br />

Nancy, France 2008.<br />

2. Al-Rubeai M, Singh RP, Goldman MH, Emery AN: Death mechanisms of<br />

animal cells in conditions of intensive agitation. Biotechnol. Bioeng 1995,<br />

45:463-472.<br />

3. Mollet M, Godoy-Silva R, Berdugo C, Chalmers JJ: Acute hydrodynamic<br />

forces and apoptosis: A complex question. Biotechnol. Bioeng 2007,<br />

98:772-788.<br />

4. Hwang SO, Lee GM: Nutrient deprivation induces autophagy as well as<br />

apoptosis in Chinese hamster ovary cell culture. Biotechnol. Bioeng 2007,<br />

99:678-684.<br />

Page 137 of 181<br />

P102<br />

Increasing antibody yield and modulating final product quality using<br />

the Freedom TM CHO-S TM production platform<br />

Michelle Sabourin 1* , Ying Huang 1 , Prasad Dhulipala 3 , Shannon Beatty 1 ,<br />

Jian Liu 1 , Peter Slade 2 , Shawn Barrett 3 , Shue-Yuan Wang 1 , Karsten Winkler 4 ,<br />

Susanne Seitz 4 , Thomas Rose 4 , Volker Sandig 4 , Peggy Lio 1 , Steve Gorfien 3 ,<br />

Laurie Donahue-Hjelle 3 , Graziella Piras 1<br />

1 Life Technologies TM , Frederick, MD, 21704, USA; 2 Life Technologies TM ,<br />

Eugene, OR, 97402, USA; 3 Life Technologies TM , Grand Island, NY, 14072, USA;<br />

4 ProBioGen AG, Goethestrasse 54, 13086 Berlin, Germany<br />

E-mail: michelle.sabourin@lifetech.com<br />

BMC Proceedings 2011, 5(Suppl 8):P102<br />

Background: Cell line development (CLD) is a critical step in the<br />

generation of biotherapeutics, but it is still hindered by several pain<br />

points, including the lengthy and labor-intensive workflow needed to<br />

isolate desirable clones, lack of reproducibility, as well as potential<br />

protein quality issues. Over the last decade, antibody titers in<br />

mammalian cell culture systems in excess of 3 g/L have been achieved<br />

through the use of novel media and feeds. However, it is still a<br />

challenge to consistently and rapidly create a stable cell line and a cell<br />

Figure 1(abstract P102) Cloning medium choice does not impact clone titer distribution. The same Molecule 1-producing stable pools were seeded for<br />

limiting dilution cloning in either a lean cloning medium prototype or CD FortiCHO TM Medium. Following clone scale-up, the top clones from each<br />

process were assessed in shake flasks using a simple fed-batch process; day 7 data are shown. While there are differences in the absolute numbers,<br />

especially for the top 3 clones, overall the clones produced similarly whether they were isolated from lean cloning medium prototype or from CD<br />

FortiCHO TM Medium.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P102) Molecule 1 clone productivity during scale-up from shake flask to bioreactor<br />

Molecule 1 clone Shake flask simple fed-batch (g/L) Shake flask fed-batch (g/L) Bioreactor fed-batch (g/L)<br />

1 0.59 0.87 2.2<br />

2 0.55 1.06 1.5<br />

3 0.52 0.96 3.3<br />

culture process capable of supporting both high antibody yield and<br />

acceptable post-translational modifications while managing the effort<br />

required for execution of the workflow. The goal of the study was to<br />

develop a robust and reproducible stable cell line workflow to generate<br />

scalable high-producing clones in less than 6 months, with industrystandard<br />

titers and desirable product quality using minimal effort.<br />

Using CHO-S as the host cell line, we first evaluated if a single medium<br />

could be used for the entire CLD workflow, therefore avoiding the issues<br />

and complications of changing media during this process. We<br />

investigated if a formulation previously shown to increase titer as a<br />

production medium could in fact be used for all CLD steps, from<br />

transfection to stable pool isolation all the way through to clone<br />

productivity, without compromising titers or performance. The same rich<br />

production medium was used in limiting dilution cloning and compared<br />

to a lean cloning medium prototype. Furthermore, robustness of the<br />

workflow was verified by testing multiple molecules. We also explored<br />

reducing effort by streamlining all the steps of the workflow. Finally, we<br />

assessed top clone scalability and expressed product quality. We tested<br />

whether clones chosen only by titers responded well to scale-up and<br />

process development in a model bioreactor setting. In addition, product<br />

glycosylation from these clones was compared to the same molecule<br />

produced in CHO DG44 cells, a well-characterized production platform.<br />

Results: Our results show that cell growth during selection and<br />

productivity assessment were affected by both the basal medium and<br />

nutrient feed strategy. Stable pools generated in either CD OptiCHO TM<br />

Medium or CD FortiCHO TM Medium gave the most desirable outcomes<br />

in terms of recovery time and productivity. We also found that CD<br />

FortiCHO TM Medium can be used for every step of the CLD workflow,<br />

including limiting dilution cloning. When we performed a limiting<br />

dilution cloning using the same pool seeded in either a lean cloning<br />

medium prototype or in CD FortiCHO TM Medium, we found that the top<br />

10 clones isolated from each medium showed similar average titer<br />

trends (Figure 1). We also established the robustness of the selection<br />

scheme by demonstrating that all tested molecules show an increase in<br />

cell pool titer during a two-stage selection scheme that uses<br />

simultaneous puromycin and methotrexate treatment. In addition, the<br />

resulting clones can readily be scaled up to bioreactors: clones that<br />

were producing ~0.5 g/L in simple fed-batch (glucose feed only) using<br />

shake flasks readily scaled-up to a DasGip bioreactor and produced up<br />

to 3.2 g/L under fed-batch mode with minimal process development<br />

(Table 1). Finally, we demonstrated that the majority of clones isolated<br />

are stable for productivity, and that the glycosylation pattern of the<br />

same molecule produced in either CHO DG44 or CHO-S TM cells was<br />

indistinguishable.<br />

Conclusions: Theuseofasinglemediumfortheentireworkflowis<br />

revolutionary, and has the distinct advantage of avoiding any need for<br />

media adaptation at any point in the workflow, whether it be preceding<br />

or following cloning, or for productivity assessment. This in turn avoids<br />

any undesirable genetic selection that may occur during such adaptation<br />

steps, and facilitates streamlining the workflow for ease of use and<br />

efficiency. Clones are easily scaled from shake flask to bioreactor and<br />

produce 2-3 g/L with minimal process development. Product<br />

glycosylation in top CHO-S TM clones was comparable to historical data<br />

from CHO DG44-derived clones expressing the same molecule. In<br />

addition, the establishment of clone stability and acceptable glycosylation<br />

patterns are key attributes required for regulatory approval of<br />

biotherapeutic production. Together, these results demonstrate the<br />

capabilities of the Freedom TM CHO-S TM Kit as an efficient and robust<br />

stable CLD platform, which can be accessed without the burden of<br />

milestone or royalty payments.<br />

Page 138 of 181<br />

The top 3 clones expressing Molecule 1 were cultured in CD FortiCHO TM Medium in shake flasks using a simple fed-batch or fed-batch process, and in a DasGip<br />

bioreactor using a fed-batch process. Clone titers increased 3- to 6-fold from simple fed-batch shake flask culture to fed-batch bioreactor culture.<br />

P103<br />

Single use bioreactors for the clinical production of monoclonal<br />

antibodies – a study to analyze the performance of a CHO cell line and<br />

the quality of the produced monoclonal antibody<br />

Sonja Diekmann * , Constanze Dürr, Alexander Herrmann, Ingo Lindner,<br />

Daniela Jozic<br />

Roche Diagnostics GmbH, Pharma Biotech Penzberg, 82377 Penzberg,<br />

Germany<br />

E-mail: Sonja.Diekmann@roche.com<br />

BMC Proceedings 2011, 5(Suppl 8):P103<br />

Background: In recent years, the use of disposables in the pharmaceutical<br />

industry has increased extensively. Disposables can be used in many areas<br />

of biopharmaceutical production. The use of disposables not only reduces<br />

investment costs, but also requires less manpower to operate, since time<br />

consuming change-over procedures are significantly reduced. In addition,<br />

disposables enable a high flexibility by reducing unit operation times such<br />

as cleaning and sterilization as well as the validation of these procedures [1].<br />

Disposable bioreactors can be subdivided into two main groups, static and<br />

dynamic systems. The dynamic systems differ with regard to the power<br />

input in stirred, vibromixed or wave-induced systems [2]. Since the power<br />

input, the mixing time, the tip speed and the oxygen transfer coefficient<br />

may influence the cell culture process and the product quality itself, a<br />

thorough characterization of the system used is necessary [3]. SSB (stainless<br />

steel bioreactors) and SUB (single use bioreactors) differ in terms of their<br />

physical design. Usually, an SSB is equipped with two or three stirrer blades.<br />

In contrast, stirred SUBs usually have just one stirrer blade. In addition,<br />

the design and the position of the stirrer differ significantly. These<br />

distinctions lead to different physical characteristics regarding the power<br />

input, mixing time, and tip speed. The SUBs are characterized by a<br />

significantly lower power input and tip speed and a significantly higher<br />

mixing time. In order to maintain a sufficient oxygen transfer coefficient, the<br />

single use bioreactor is equipped with a micro sparger with a pore size of<br />

25 µm. Furthermore, pure oxygen can be used to achieve higher dissolved<br />

oxygen concentrations in the cell cultivation medium. This study describes<br />

the influence of these technical differences on the performance of a CHO<br />

cell line and the product quality of a monoclonal antibody.<br />

Results: After inoculation, the cells were cultured in both systems in a fed<br />

batch mode with two continuous feeds for twelve days. Figure 1a shows the<br />

viable cell density as a function of time. Whereas the overall growth<br />

characteristic of the cells in both systems are comparable over the whole<br />

cultivation time, from 92 h until 188 h, the cells in the SSB are characterized<br />

by a significantly (*) faster growth (p< 0.05). The viability of the cells in both<br />

systems remains between 90% and 100% over the whole cultivation time<br />

(data not shown). Figure 1b shows the LDH activity in both systems as a<br />

function of time as an indicator of cell lysis. Up to 68 h cultivation time the<br />

LDH activity in both systems is comparable. Thereafter, the LDH activity<br />

measured in the SUB is significantly (*) higher in comparison to the SSB (p <<br />

0.05). Figure 1c shows the product titer [%] produced by the cells in the SSB<br />

in comparison to the SUB and Figure 1d shows the specific productivity.<br />

Between 140 h and 250 h the product titer in the SSB is slightly higher in<br />

comparison to the SUB, whereas the titer at harvest is comparable. Since the<br />

growth of the cells in the SSB is faster in comparison to the cells in the SUB,<br />

the specific productivity of the cells cultivated in the SUB is higher. Figure<br />

1d shows the IEC data of the antibody produced by the CHO cells in the<br />

SUB and the SSB. Whereas the acidic region of the antibody of the SSB is<br />

slightly higher in comparison to the antibody of the SUB the difference is<br />

not significant. The main peaks are comparable. The basic region of the<br />

antibody of the SSB is slightly lower in comparison to the SUB, balancing


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1a(abstract P103) Viable cell density.<br />

Figure 1b(abstract P103) LDH activity.<br />

Figure 1c(abstract P103) Product titer.<br />

Page 139 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1d(abstract P103) IEC pattern.<br />

Table 1(abstract P103) SEC pattern<br />

Bioreactor-type Monomer<br />

[%]<br />

SSB 99.7 ± 0 0.267 ± 0.047 0.1 ± 0<br />

SUB 99.7 ± 0 0.3 ± 0 0.1 ± 0<br />

Figure 1e(abstract P103) Glycopattern.<br />

the slight increase in acidic region observed for the antibody derived from<br />

the SSB.<br />

The SEC patterns of both products are almost identical (Tab. 1). The<br />

glycopatterns of the mAB produced in the SUB and of the mAB produced<br />

in the SSB shows no significant differences (Fig. 1e). The G0 fraction of the<br />

mAB produced in the SSB is slightly higher in comparison to the G0<br />

fraction of the mAB produced in the SUB whereas the G1 fraction of the<br />

mAB produced in the SSB is slightly lower compared the mAB produced in<br />

the SUB. The G2 fraction is very similar. The G0-Fucose value of the mAB<br />

produced in the SSB is higher than for mAB produced in the SUB, which<br />

leads to a higher overall a-fucose value for the SSB derived product<br />

compared to the SUB derived product. However, all these differences are<br />

not significant. All other fractions are comparable between both<br />

antibodies. To investigate the influence of both bioreactor types on the<br />

impurity profile, the DNA and HCP values of the harvested supernatant<br />

were compared. Figure 1f shows the specific DNA concentration (DNA<br />

HMW<br />

[%]<br />

Page 140 of 181<br />

LMW<br />

[%]<br />

concentration divided by viable cell density) of the harvested supernatant<br />

of the SSB and the SUB. The specific DNA concentration of the harvested<br />

supernatant of the SSB is slightly higher compared to the SUB. However,<br />

theses differences are not significant. Figure 1g shows the specific HCP<br />

concentration measured in the harvested supernatant of the SSB and the<br />

SUB. The specific HCP concentration (HCP concentration divided by viable<br />

cell density) of the SUB is slightly higher compared to the specific HCP<br />

concentration of the SSB. Again, these differences are not significant.<br />

Conclusions: The present study describes the influence of a single use<br />

bioreactor on the performance of a production CHO cell line, on the<br />

product quality of the produced antibody and the process related<br />

impurities in comparison to a commercial stainless steel bioreactor. It<br />

seems that the microsparger of the SUB lead to a cell damage, which is<br />

measured by LDH activity. Nevertheless, our findings indicate that this<br />

cell damage has no influence on the productivity, the concentration of<br />

DNA and HCP and, most importantly on the quality of the antibody.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1f(abstract P103) DNA concentration.<br />

Figure 1g(abstract P103) HCP concentration.<br />

References<br />

1. Brecht R: Disposable bioreactors: maturation into pharmaceutical<br />

glycoprotein manufacturing. Adv Biochem Eng Biotechnol 2009, 115:1-31.<br />

2. Eibl R, Eibl D: Application of Disposable Bag Bioreactors in Tissue<br />

Engineering and for the Production of Therapeutic Agents. Adv Biochem<br />

Eng Biotechnol 2008.<br />

3. Hanson MA, Brorson KA, et al: Comparisons of optically monitored smallscale<br />

stirred tank vessels to optically controlled disposable bag<br />

bioreactors. Microb Cell Fact 2009, 8:44.<br />

4. Hacker DL, De Jesus M, et al: 25 years of recombinant proteins from<br />

reactor-grown cells - where do we go from here? Biotechnol Adv 2009,<br />

27(6):1023-1027.<br />

P104<br />

Development of live cultural pandemic influenza vaccine Vector-Flu<br />

Elena A Nechaeva 1* ,TatyanaYSen’kina 2 , Alexander B Ryzhikov 2 , Irina F Radaeva 2 ,<br />

Ol’ga G P’yankova 2 ,Natal’ya V Danil’chenko 2 , Tatyana M Sviridenko 2 ,<br />

Marina P Bogryantzeva 2 ,Natal’ya V Gilina 2 , Nikolay A Varaksin 2 ,<br />

Tatyana G Ryabicheva 2 , Irina V Kiseleva 3 , Larisa G Rudenko 3<br />

1 Department of Cell Culture Technology, SRC VB VECTOR, Novosibirsk, 630559,<br />

Russia; 2 SRC VB VECTOR, Novosibirsk, 630559, Russia; 3 Institute of Experimental<br />

Medicine, Russian Academy of Medical Sciences, St. Petersburg, 197376, Russia<br />

E-mail: nechaeva@vector.nsc.ru<br />

BMC Proceedings 2011, 5(Suppl 8):P104<br />

Page 141 of 181<br />

Background: The threat of pandemic A/H1N1 influenza remains a matter<br />

of considerable public concern. Recent influenza outbreaks underline the<br />

importance of rapid production of a reserve of vaccine sufficient for<br />

pandemic and interpandemic periods. Traditional vaccines intended for<br />

seasonal influenza do not satisfy this demand, because they are unable to<br />

induce cross-reactive antibodies to pandemic flu.<br />

Live influenza vaccines made in the cell culture offer potential advantages<br />

because it is possible to produce a considerable amount of these vaccines<br />

over a short period. Their use eliminate concerns about allergic reactions to<br />

egg proteins, and they allow the antigenic composition of the vaccine to be<br />

closer to the circulating influenza virus strains.<br />

The goal of this work was to develop a live attenuated vaccine against<br />

pandemic influenza, Vector-Flu, derived from cold-adapted strain A/17/<br />

California/2009/38(H1N1) and to study the immunogenic and protective<br />

properties of the vaccine using ferrets and mice.<br />

Materials and methods: Cell culture: MDCK from Cell Culture Collection<br />

of SRC VB VECTOR. Cells were passed in serum-free SFM4MegaVir<br />

medium (USA). The characteristics of the MDCK cell line were studied in<br />

accordance with WHO [1].<br />

Viruses: The vaccine strain A/17/California/2009/38(H1N1) was generated<br />

at the Institute of Experimental Medicine (St. Petersburg, Russia) by<br />

reassortment of the cold-adapted attenuated A/Leningrad/134/17/57<br />

(H2N2) master donor virus with the pandemic strain A/California/7/2009<br />

(H1N1). The A/Chita/3/2009(H1N1) influenza virus was obtained from<br />

VECTOR’s Collection of Microorganisms.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P104) The titers of ferret blood serum to A/H1N1 influenza virus strains before and after<br />

immunization with the Vector-Flu vaccine determined by HAI and microneutralization assay<br />

Serological method HAI Microneutralization<br />

Influenza virus strain A/Chita/3/2009 A/California/7/2009 A/Chita/3/2009 A/California/7/2009<br />

Number of vaccinations 0 1 2 0 1 2 0 1 2 0 1 2<br />

GMT a over the group


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P105) Viable cell density (●, ○), viability (▲, △) and perfusion rate (■, □) in perfusion processes using ATF system (A) and TFF system (B).<br />

Closed symbols indicate the ATF9 perfusion experiment, whereas the open symbols represent the ATF15 and TFF10 perfusion experiments.<br />

Supplementations of glucose or glutamine were performed according to cell<br />

need. The pH was controlled by adding 0.5 M Na 2CO 3 or pulsing CO 2 into<br />

the headspace. The cell density, viability, pH, pCO2, concentrations of<br />

glucose, lactate, glutamine and ammonia were measured by Bioprofile FLEX<br />

(Nova Biomedical). Antifoam C (Sigma Aldrich, US) was added up to 50 ppm<br />

concentration in the bioreactor either by boost addition or by continuous<br />

pumping. The mAb concentration was measured by protein A HPLC.<br />

Results: Cell density, viability and perfusion rate: Two perfusion<br />

experiments (ATF9 and ATF15) were conducted using ATF (Figure 1A) and<br />

one perfusion culture (TFF10) using TFF device (Figure 1B). In experiments<br />

ATF9 and TFF10, cell density was maintained between 20-30 x 10 6 cells/<br />

mL by daily cell bleeds for 2 weeks using one HF. Interestingly, in<br />

experiment ATF9, the use of an ATF flow rate of 0.7 L/min was not<br />

sufficient to remove bubbles entrapped in the HF resulting in a decrease<br />

in viability (93.5% vs. 97%). Therefore, from day 9, the flow rate was<br />

increased to 1 L/min, i.e. shear stress of 3400 s -1 . Experiment ATF15 was<br />

performed at 1 L/min.<br />

In experiment ATF15, a cell density of 132 x 10 6 cells/mL was reached after<br />

10 days of culture of exponential growth. Reaching this density coincided<br />

with interruption of ATF function due to insufficient pressure to push the<br />

highly viscous cell broth, showing the limit of this system at this very high<br />

cell density when using a non-pressurisable disposable bioreactor. A batch<br />

culture was then carried out between days 13 and 20 (data not shown)<br />

after which perfusion was re-started. A maximal cell density of 123 x 10 6<br />

cells/mL was obtained confirming the first observed maximal cell density.<br />

The culture was then continued for 4 days at cell density around 100 x 10 6<br />

cells/mL by performing daily cell bleeds showing that healthy high cell<br />

density culture could be obtained somewhat under 130 x 10 6 cells/mL in<br />

the same culture.<br />

Using the TFF, exponential cell growth was obtained at similar rate as with<br />

ATF, the cell density was then maintained around 100 x 10 6 cells/mL by<br />

daily cell bleeds for 2 weeks. Then, higher cell densities were achieved, up<br />

to more than 200 x 10 6 cells/mL for 2 days. Reaching 214 x 10 6 cells/mL<br />

coincided with the limit of this system due to high pressure in re-circulation<br />

Table 1(abstract P105) Comparison of mAb production using ATF or TFF system at day 17 with cell densities<br />

maintained at 20-30 x 10 6 cells/mL<br />

ATF TFF<br />

Production in harvest (g) 9 7<br />

Yield (production in harvest/total production) (g/g in %) 75 55<br />

Total removal of mAb in cell bleeds/total production (g/g in %) 19 30<br />

Residual mAb mass in bioreactor/total production (g/g in %) 6 15<br />

Page 143 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

loop (1 bar) related to the viscosity, oxygenation limitation and CO 2<br />

accumulation (31 kPa). During these runs, CHO cells were cultivated at high<br />

and very high cell densities with a cell viability maintained above 90%<br />

(mostly around 95%).<br />

Total mAb production: Over an 17-day period of culture with cell<br />

densities maintained at 20-30 x 10 6 cells/mL, a total of 9 g and 7 g of mAb<br />

were harvested in the ATF9 and TFF10 processes respectively (Table 1).<br />

The lower yield obtained using the TFF was mainly due to mAb removed<br />

in cell bleeds (30% in TFF vs. 19% in ATF). In both cultures, the cell specific<br />

production rates of mAb were similar and around 10-15 pg/cell/day (data<br />

not shown).<br />

Conclusions: A very high cell density of 100 x 10 6 cells/mL was stably<br />

maintained in growing phase and at high viability by performing cell<br />

bleeds in a perfused WAVE Bioreactor using TFF or ATF cell separations.<br />

With the settings used here, the maximal cell density was limited to<br />

214 x 10 6 cells/mL using TTF and 132 x 10 6 cells/mL using ATF. Using<br />

TFF, the cell density could not be further increased due to the limitations<br />

of membrane capacity (for the encountered high viscosity), oxygenation<br />

and CO 2 level, and using ATF due to pressure limitation to push highly<br />

viscous fluid and use of non-pressurisable disposable bioreactor. Thus, the<br />

TFF system allowed reaching higher cell densities. To our knowledge, it is<br />

the first time that a CHO cell density of more than 200 x 10 6 cells/mL<br />

was achieved in a wave-agitated bioreactor.<br />

Higher retentions of mAb were observed using the TFF system than using<br />

the ATF system. In perfusion, the major effect of this retention was loss of<br />

mAb in the cell bleeds. Consequently, ATF system was more favourable<br />

for production at stable cell density maintained by cell bleeds.<br />

The use of a disposable bioreactor equipped with a disposable separation<br />

device offers a solution alleviating technical and sterility challenges<br />

occurring in perfusion processes.<br />

Acknowledgements<br />

This work was financed by GE Healthcare. Thank you to Eric Fäldt,<br />

Christian Kaisermayer and Craig Robinson from GE Healthcare. We<br />

acknowledge The Swedish Governmental Agency for Innovation Systems<br />

(VINNOVA), which supported the lab activity. We thank IMED, Sweden, for<br />

their kind permission of using their research cell line. We would like also<br />

to acknowledge Refine Technology for helpful inputs.<br />

P106<br />

Utilizing Roche Cedex Bio analyzer for in process monitoring in biotech<br />

production<br />

Dörthe Druhmann * , Sabrina Reinhard, Felizitas Schwarz, Christina Schaaf,<br />

Katrin Greisl, Tim Nötzel<br />

Roche Diagnostics GmbH, Pharma Biotech Penzberg<br />

BMC Proceedings 2011, 5(Suppl 8):P106<br />

Introduction: One task during development and control of bioprocesses<br />

used for production of recombinant proteins is to deliver fast, accurate<br />

and reliable analytical process data.<br />

Monitoring of nutrients and metabolites in animal or bacterial cell culture is<br />

essential to avoid limitations or toxic accumulation during fermentations<br />

that can effect cell growth, viability, product yield and quality. Within certain<br />

fermentation processes low levels of nutrients trigger in-process actions<br />

such as feed. Other parameters are used to track culture reproducibility and<br />

to gain process understanding for process development and validation.<br />

Typical parameters in mammalian cell culture are glucose, lactate,<br />

glutamine, glutamate, ammonia, sodium and potassium. Therefore it is<br />

common to use multi-analyzer systems based on enzymatic-dependent<br />

biosensors and ion- selective potentiometry.<br />

Furthermore the release of lactate dehydrogenase (LDH) into the<br />

fermentation media is used as an indicator of cell death during fermentation<br />

processes therefore a 96-well based photometric assay is used . The<br />

measurement of product titer like IgG is needed to track productivity or<br />

yield during fermentation and protein purification processes and is mostly<br />

done by HPLC methods.<br />

One major motivation for this new Bioprocess analyzer lies within the<br />

eminent issues of enzyme membrane based analytical technology such<br />

as: short life cycles as well as poor or changing quality of the enzyme<br />

membranes, high material costs, non-linearity of measurements and in<br />

particular insufficient sensitivity and accuracy.<br />

Additionally the laborious maintenance required to operate different<br />

analytical devices and labor intensive sample management as well as<br />

manual steps to avoid operator dependent variability needed to be reduced.<br />

Applying diagnostic knowledge to bioprocess care: Combining the<br />

knowledge of 25 years of diagnostic healthcare with expert knowledge in<br />

fermentation processes a new Bioprocess analyzer (Cedex Bio analyzer*) was<br />

developed. The new Cedex Bio analyzer stands out with increased sensitivity<br />

plus enlarged measurement ranges compared to currently used methods<br />

and analyzers.<br />

An automated dilution increases the measurement ranges and reduces<br />

operator variability because no manual dilution is needed before<br />

applying the sample to the Cedex Bio analyzer.<br />

The aim of this evaluation and method validation was to compare this<br />

new device to common analytical systems.<br />

Validation and correlation studies: Reference standards were analyzed<br />

at least once a day before and after each tray of samples to control the<br />

performance of the membrane analyzer. Day to day and side by side<br />

comparability as well as accuracy and linearity of the Cedex Bio analyzer are<br />

superior to the membrane analyzer as shown in Figure 1.<br />

An increased sensitivity allows to operate nutrient limited fermentations<br />

and likewise to detect remote changes within the metabolite profile of<br />

the fermentation process.<br />

Monitoring of fermentations processes using a membrane analyzer<br />

required laborious maintenance, a lot of calibrations and permanent<br />

control of recovery.<br />

However correlation studies conducted with samples of a fed-batch<br />

mammalian cell fermentation show that the Cedex Bio analyzer easily<br />

replaces the membrane analyzer with comparable batch data (Figure 2).<br />

Furthermore additional parameters like LDH or IgG can be analyzed from<br />

the same sample.<br />

Summary: ο The Cedex Bio analyzer combines three devices to one with<br />

the possibility to measure up to 9 parameters in fermentation browth at<br />

once and from the same sample within a few minutes.<br />

ο Automated dilution reduces operator dependent manual steps and<br />

variability.<br />

Table 1(abstract P106) measurement ranges current methods versus Cedex Bio<br />

Parameter Current methods Cedex Bio Cedex Bio<br />

w/o dilution automated dilution<br />

glucose [mg/L] 200 – 15.000 1)<br />

20 – 7.500 20 – 75.000<br />

lactate [mg/L] 200 – 5.000 1)<br />

18 – 1.400 18 – 14.000<br />

ammonia [mg/L] 3,6 – 450 1)<br />

0.48 – 12 0.48 – 480<br />

glutamine [mg/L] 30 – 877 1)<br />

60 – 1.500 60 – 7.500<br />

glutamate [mg/L] 30 – 882 1)<br />

15 – 1.500 15 – 7.500<br />

sodium [mg/L] 920 – 5.006 2)<br />

460 – 5.750 460 – 5.750<br />

potassium [mg/L] 39,1 – 978 2)<br />

39 – 1.170 39 – 1.170<br />

LDH [U/L] 20 – 150 3)<br />

20 – 1.000 20 – 10.000<br />

IgG [mg/L] 35 – 1.500 4)<br />

10 – 1.600 10 – 16.000<br />

1) Membrane analyzer, 2) Ion selective electrode 3) 96-well-plate assay, 4) HPLC method.<br />

Page 144 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

ο Methods based on wet chemical photometric assays, ionselective<br />

electrodes and turbidity assays are easy to handle, reliable and<br />

robust.<br />

ο The compact analyzer helps to save laboratory space and costs for<br />

different instruments.<br />

ο Sensitive, precise and accurate analytical data allow a tight control of<br />

cell culture processes.<br />

ο The assay portfolio will also be suitable for microbial fermentation with<br />

additional parameters like acetate or ethanol.<br />

ο The Cedex Bio analyzer combines three devices to one with the<br />

possibility to measure up to 9 parameters in fermentation browth at once<br />

and from the same sample within a few minutes.<br />

* CEDEX is a trademark of Roche<br />

Page 145 of 181<br />

Figure 1(abstract P106) Comparison of recovery values of the Cedex Bio analyzer versus a membrane analyzer. Range of recovery of reference standards<br />

(quality control material, n ~ 250, t = 6 months).<br />

Figure 2(abstract P106) Field Data with samples from mammalian cell culture: Cedex Bio analyzer versus a membrane analyzer. The slope < 1 due to a<br />

constant overestimation of the glucose concentration by the membrane analyzer.<br />

P107<br />

LDH-C can be differentially expressed during fermentation of CHO cells<br />

Berthold Szperalski 1* , Christine Jung 1 , Zhixin Shao 1 , Anne Kantardjieff 2,3 ,<br />

Wei-Shou Hu 2<br />

1 Pharma Biotech, Roche Diagnostics GmbH, 82377 Penzberg, Germany;<br />

2 University of Minnesota, Minneapolis, MN 55455, USA; 3 Alexion<br />

Pharmaceuticals, Cheshire, CT 06410, USA<br />

E-mail: berthold.szperalski@roche.com<br />

BMC Proceedings 2011, 5(Suppl 8):P107<br />

Methods: CHO-cells producing a recombinant human antibody were<br />

cultivated in a proprietary proteinfree medium and inoculated in 4 x 2L


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P107)<br />

Correlation to PC Gene set Number of genes in gene set Nominal p-value<br />

Positive to PC 1 Cell cycle 26 0<br />

DNA replication 24 0<br />

Cytoskeleton 67 0.02<br />

Microtubule organizing center 24 0.04<br />

Negative to PC 1 Golgi apparatus 50 0<br />

Cell-cell signaling 48 0<br />

Positive to PC 2 RNA processing 43 0.01<br />

Proteolysis 46 0.05<br />

Negative to PC 2 DNA replication 32 0.04<br />

stirred tank bioreactors. Bioreactors were controlling pH, pO2 and<br />

temperature. A fixed feeding protocol was used to overcome the<br />

limitation of consumed medium components. Temperatures of 2 cultures<br />

were shifted at day 4 from 37°C to 34°C. Daily samplings of the cultures<br />

were performed to monitor cell density and viability by using an<br />

automated Cedex cell counter and the trypan blue exclusion method.<br />

The supernatant of the culture was monitored for product concentration,<br />

glucose, glutamine, lactate, ammonium. Measurement of LDH (lactate<br />

Figure 1(abstract P107)<br />

Page 146 of 181<br />

dehydrogenase ) in cell culture supernatant was used as an indicator of<br />

cell lysis. Sedimented cells of cell culture samples were prepared and<br />

cRNA was processed according to Affymetrix standard procedures.[1]<br />

and hybridized with custom CHO Affymetrix arrays from the<br />

Consortium for Chinese Hamster Ovary (CHO) Cell Genomics [2].<br />

Results: The comparison of temperature shifted and control cultures<br />

showed significant differences in the growth curves of the experiment.<br />

Temperature shift induced an early shift to the plateau phase. It reduced


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

the cell death. Cell specific productivity was slightly higher. Lactate<br />

consumption was higher and started earlier than in control cultures (data<br />

not shown). PCA (principal component analysis) was used to compare<br />

expression ratios at different temperatures. PC 1 showed that most<br />

expression changes are onset at day 6 and maintained throughout the<br />

rest of the culture. Transcriptome analyses showed several significant<br />

changes after the temperature shift (Table 1). One outstanding result is<br />

the upregulated RNA of LDH-C (Figure 1). LDH-A RNA expression showed<br />

no significant change after temperature shift.<br />

Discussion: LDH-C is known to be present in sperm cells , testis cells<br />

and some tumors [3] but is not reported to be regulated in CHO-cell<br />

lines. In sperm cells LDH-C is known to have different kinetic properties<br />

compared to A and B isoforms of LDH preferring lactate as substrate<br />

[4]. LDH-C is localized in cytoplasm and in specific “sperm type<br />

mitochondria” and seems to be integrated in a shuttle system for the<br />

transfer of reducing activity into the mitochondrial matrix [7][8]. An<br />

pseudogene association with mitochondrial cyclophilin D is reported in<br />

thegenebankofmousegenome[9].TheroleofLDH-CinCHO-Cellsis<br />

still unclear. The influence of temperature shift under normal body<br />

temperature seems to induce a special situation for sperm cell<br />

migration. LDH-C helps sperm cells to survive in lactic acid containing<br />

micro milieus of the oviduct. It allows lactic acid to be an energy<br />

source. These functions could be mimicked in a high lactate containing,<br />

temperature shifted fermentation process with CHO cells. LDH-C can<br />

also be regulated by hormonal mechanisms. They are known to have<br />

slight regulatory influence on the transcriptional expression [5].<br />

Selective inhibitors of LDH isoforms are described [6]. Specific inhibitors<br />

for LDH -C are proposed as antifertilizing drugs [6]. Inhibitors to LDH-A<br />

and -B could help to favor LDH-C and so reduce lactate production.<br />

LDH-C is an interesting target for engineering manufacturing processes<br />

with cell lines like CHO cells for shifting these cells to aerobic lactate<br />

metabolism and improving growth performance.<br />

References<br />

1. Kantardjieff A, Jacob NM, Yee JC, Epstein E, Kok YJ, Philp R, Betenbaugh M,<br />

Hu WS: Transcriptome and proteome analysis of Chinese hamster ovary<br />

cells under low temperature and butyrate treatment. J Biotechnol 2010,<br />

145:143-159.<br />

2. Consortium for Chinese Hamster Ovary (CHO) Cell Genomics. 2007<br />

[http://hugroup.cems.umn.edu/CHO/cho_index.html].<br />

3. Koslowski M, Tureci O, Bell C, Krause P, Lehr HA, Brunner J, Seitz G,<br />

Nestle FO, Huber C, Sahin U: Multiple splice variants of lactate<br />

Figure 1(abstract P108) Curves, reflecting change of probability, versus stiffness parameter, at different values of k.<br />

Page 147 of 181<br />

dehydrogenase C selectively expressed in human cancer. Cancer Res<br />

2002, 62:6750-6755.<br />

4. Blanco A, Burgos C, Gerez de Burgos NM, Montamat EE: Properties of the<br />

testicular lactate dehydrogenase isoenzyme. Biochem J 1976,<br />

153:165-172.<br />

5. Ohsako S, Kubota K, Kurosawa S, Takeda K, Qing W, Ishimura R, Tohyama C:<br />

Alterations of gene expression in adult male rat testis and pituitary<br />

shortly after subacute administration of the antiandrogen flutamide. J<br />

Reprod Dev 2003, 49:275-290.<br />

6. Yu Y, Deck JA, Hunsaker LA, Deck LM, Royer RE, Goldberg E, Vander<br />

Jagt DL: Selective active site inhibitors of human lactate dehydrogenases<br />

A4, B4, and C4. Biochem Pharmacol 2001, 62:81-89.<br />

7. Burgos C, Maldonado C, Gerez de Burgos NM, Aoki A, Blanco A:<br />

Intracellular localization of the testicular and sperm-specific lactate<br />

dehydrogenase isozyme C4 in mice. Biol Reprod 1995, 53:84-92.<br />

8. Gladden LB: Lactate metabolism: a new paradigm for the third<br />

millennium. J Physiol 2004, 558:5-30.<br />

9. Mouse DNA sequence from clone RP23-313I15 on chromosome 13<br />

Contains a peptidylprolyl isomerase D (cyclophilin D) (Ppid) pseudogene<br />

and a sperm specific lactate dehydrogenase 3C (Ldh3) pseudogene,<br />

complete sequence. GenBank: AL606965.20 2011 [http://www.ncbi.nlm.<br />

nih.gov/nucleotide/AL606965.20].<br />

P108<br />

Stem cell: from basic theoretical assumptions and mathematical<br />

concepts to the computational models<br />

Katya Simeonova 1* , Ganka Milanova 2<br />

1<br />

Institute of Mechanics, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;<br />

2<br />

University of Architecture, Civil Engineering and Geodesy, 1000 Sofia,<br />

Bulgaria<br />

E-mail: katyas@bas.bg<br />

BMC Proceedings 2011, 5(Suppl 8):P108<br />

Background: Stem cells (SC) and their therapeutic applications have a<br />

great promise for treatment of many human diseases, [1]. Characteristics<br />

of Embryonic Stem Cells (ESC) have been presented in [2]. The aim of<br />

the paper presented has been formulated as follows: to give some<br />

classical mechanics and mathematics theories for SC studies.<br />

Mathematical models for cancer diseases and neurological disorders have<br />

been discussed too.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Materials and methods: Models, including a viscoelastic continuum, a<br />

combination of viscoelastic fluid have been developed as well, [3]. By<br />

relatively modern (for that time) microscopic techniques, developed<br />

during the eighteenth have been observed the images of living cells.<br />

Later using imaging mechanics, have been discovered AFM, imaging for<br />

study of living cells. High resolution of AFM imaging “has provided<br />

information on the structure. It has been proved as well, [4] that blood,<br />

blood vessels and nerves, could be tested mechanically.<br />

Mathematical models of SC for neuroscience: Theoretical concepts<br />

and mathematical models have been proposed for explanation<br />

quantitatively biological mechanisms. Mechanisms and models of<br />

cellular State Transition have been discussed too. Other periodic<br />

hematological diseases involve oscillations in all of the blood cells. In<br />

the paper [5], has been developed a simple mathematical model0. The<br />

probabilities for CSC to reach levels, compatible with the diagnosis of<br />

acute leukemia, by CSC, as well as probability for extinction have been<br />

computed analytically.<br />

An analytical model: The probability “that at least once, the population<br />

will have M 1CSC, approximately given by:<br />

1 r<br />

pM ( 0, M1)<br />

=<br />

1 r<br />

−<br />

−<br />

−M0<br />

−M<br />

Here M 1CSC, at time t =0,r is the relative fitness parameter of CSC. The<br />

general expression for the fixation probability could be given as follows:<br />

( ) =<br />

p M , M<br />

0 1<br />

q<br />

q<br />

( M0<br />

)<br />

( M )<br />

1<br />

For the birth-exports process authors in [11], considered the transition<br />

probability T*(k, NSC ), that numbers of CSC increasing from k to k+1,<br />

given by formula:<br />

p k<br />

CSC ( ) =<br />

kr<br />

kr + N − k<br />

( )<br />

A computational model, results: Solving the equation (4) we obtained<br />

the next formula for k:<br />

pN<br />

k =<br />

p + r − pr<br />

Here all parameters: p, r and N have been presented in the work [5]. So<br />

we investigate the effect of parameter k, on the probability p CSC(k).<br />

Numerical FORTRAN programs designed by authors have been used. By<br />

numerical experiments have been obtained dependencies at different k.<br />

Results, have been analyzed and shown (Figure 1).<br />

Conclusions: Future problems and current studies on SC are: SC is very<br />

promising tool for various biopharm applications, Future technologies<br />

will be enabled fuller understanding of SC, Future directions for human<br />

S Cell Culture optimization, ES could be used as vechile for gene<br />

therapy.<br />

Acknowledgments: We thank you to Professor Nicole Borth, and to<br />

Organizers of ESACT2011, for their great cooperation to attend ESACT<br />

Meeting in Vienna, 15-18 May, 2011 and to present both poster reports.<br />

References<br />

1. Yanhong S: Stem Cell Research and therapeutics: Advances in<br />

Biomedical Research. SPRINGER: Deunis O 2008.<br />

2. Simeonova K: Nucleic Acids Res. ESF -UB Conference Rare Diseases II:<br />

Hearing and Sight Loss Costa Brava, Spain 2009, 105[http://www.esf.org/<br />

conferences/09295].<br />

3. Satin BD, et al: Nucleic Acids Res.32:4876-4879.<br />

4. Ingo R, Radtke F: Stem cell biology meets system biology. Meeting Review,<br />

Development 136 2009, 3525-3530.<br />

5. Dingli D, et al: Stochastic dynamics of hematopoietic tumor stem cells.<br />

Cell Cycle 2007, 6:e1-e6.<br />

(1)<br />

(2)<br />

(3)<br />

(4)<br />

Page 148 of 181<br />

P109<br />

Comparison of the activity and pluripotency maintaining potential of<br />

human leukemia inhibitory factor (LIF) produced in E.coli and CHO cells<br />

Claas Haake 1 , Sophia Bonk 1 , Jana Parsiegla 1 , Magda Tomala 1 , Komal Loya 2 ,<br />

Malte Sgodda 2 , Tobias Cantz 2 , Axel Schambach 3 , Cornelia Kasper 1 ,<br />

Thomas Scheper 1*<br />

1 Institute for Technical Chemistry, Leibniz University of Hanover, 30167<br />

Hannover, Germany; 2 Junior Research Group Stem Cell Biology, Cluster of<br />

Excellence REBIRTH; MPI Münster and Hanover Medical School, Hannover<br />

Germany; 3 Department of Experimental Hematology, Hanover Medical<br />

School, 30625 Hannover, Germany<br />

E-mail: scheper@iftc.uni-hannover.de<br />

BMC Proceedings 2011, 5(Suppl 8):P109<br />

Introduction: Leukemia inhibitory factor (LIF) is a polyfunctional cytokine<br />

with numerous regulatory effects in vivo and in vitro. In murine stem cell<br />

cultures it is the essential media supplement for the maintenance of<br />

pluripotency of embryonic and induced pluripotent stem cells. To explore<br />

if the glycosylation and/or other post-translational modifications are<br />

affecting this activity, we produced and isolated LIF from either<br />

eucaryotic cells (Chinese hamster ovary (CHO) cells) or procaryotes (E.coli)<br />

and compared their biological activities in this study.<br />

Results:<br />

Production in E.coli: To increase the solubility the LIF is expressed<br />

together with thioredoxin as a fusion protein. The genes for the fusion<br />

protein are separated on the vector (pET32b) by a TEV (Tobacco Etch<br />

Virus) protease cleavage site, in addition thioredoxin is expressed with a<br />

his-tag. The resulting construct was transformed into E.coli BL21(DE3),<br />

which were afterwards cultivated at 23°C. This relatively low temperature<br />

leads to increased solubility and yield of the target protein. The protein<br />

was afterwards purified from the fermentation broth by metal chelate<br />

affinity chromatography through the utilization of Zn 2+ ions immobilized<br />

on IDA membrane adsorbers. To cleave the thioredoxin from the fusion<br />

protein the obtained protein fractions were directly treated with<br />

recombinant TEV protease. The released hLIF was purified from the<br />

protein mixture using ion exchange chromatography [1].<br />

Production in CHO cells: The utilized CHO SFS cell line (CCS, Hamburg,<br />

Germany) was transducted using a lentivirus with a pRRL vector, which<br />

coded for his-tagged humane LIF. Verification of the expression, as well<br />

as of the extracellular localization was carried out by intracellular flow<br />

cytometric staining and by western blot.Thecultivationinserumfree<br />

medium (ProCHO5, Lonza, Basel, Switzerland) was initially performed in<br />

spinner flasks. Afterwards the scale-up to 1.5 L bioreactor scale was<br />

accomplished. The cultivation was carriedoutat37°C,200rpm,andpH<br />

7.2. The first step of the purification of the concentrated supernatant was<br />

performed by metal chelate affinity chromatography, where Ni 2+ ions<br />

were immobilized. Afterwards the pooled protein fractions were purified<br />

by a polishing step by heparin affinity chromatography.<br />

Effects on murine iPS suspension cells: Cell growth: The cells were<br />

cultivated as cell spheres in suspension in DMEM medium supplied with<br />

10 ng LIF per ml. As positive control ESGRO LIF, which is produced in<br />

E.coli, was used. For each of the three LIFs used as well as for the<br />

negative control two cultures of 2 ml each were harvested using trypsin<br />

every day and the cell count was determined manually using trypan blue.<br />

For the long-term cultivation the cells were passaged twice a week. The<br />

obtained values for maximal cell density and the viabilities show the<br />

activity of the produced LIFs when compared with the positive and<br />

negative control but no discrepancy in their activities.<br />

>Pluripotency maintaining potential: Via flow cytometry the expression of<br />

the pluripotency markers SSEA-1 and Oct4 was determined using an anti-<br />

SSEA-1-PE antibody and measuring the expression of GFP, which stands<br />

under the control of an Oct4 promoter respectively. For the negative<br />

control an isotype control antibody was used for SSEA-1. For GFP, the<br />

results achieved were compared to the GFP expression of the cells at the<br />

start of the cultivation. The results show a slight decrease in SSEA-1 (FL-2)<br />

but not GFP (FL-1) expression over the duration of the cultivation, but no<br />

differences regarding the activities in accordance to the source of the LIFs.<br />

Effects on adherent growing murine ESC: Murine ESC, carrying an<br />

Oct4-GFP reporter construct (OG2-ESC) [2] were seeded on C3H irridatred<br />

mouse embryonic fibroblast cells in 6-well-plates with LIF concentrations<br />

of 40, 10, and 2.5 ng/ml. As positive control an E.coli-derived LIF was


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P109) Living cell count and viability against the cultivation time for the cultivation of the ES cells with 10 ng LIF/ml.<br />

used, which has been produced at the MPI Münster and has previously<br />

been tested. The media were changed every 2 days and the cells were<br />

passaged twice a week for 5 passages. After 24 days the cells were<br />

harvested and used for qRT-PCR. Within the qRT-PCR b-Actin was used as<br />

the housekeeping gene. The expression of the pluripotency markers Oct4<br />

and Nanog as well as marker proteins for the three germ layers (Trp63<br />

for ectodermal, AFP for endodermal and Brachyury for mesodermal<br />

differentiation) were quantified. The results show that the mesodermal<br />

marker is downregulated, which indicates spontaneous differentiation of<br />

the negative control into the mesodermal germ layer. Between the used<br />

LIFs no significant differences could be determined.<br />

Effects on murine ES suspension cells: Cell growth: The procedure was<br />

in accordance to the approach with the iPS cells and the three LIFs were<br />

also used in concentrations of 100 and 1 pg each. Supplementary the<br />

amount of apoptosis was measured after four days of culture using the<br />

Annexin-V Kit (Invitrogen, Carlsbad, CA, USA) for flow cytometry. The results<br />

clearly show activity for the concentrations of 10 ng (figure 1) and 100 pg<br />

but no considerably difference from the negative control for the<br />

concentration of 1 pg and also no differences for the LIFs regarding their<br />

sources were found.<br />

Pluripotency maintaining potential: ThemarkerproteinSSEA-1was<br />

measured flow cytometrically once a week for the culture containing<br />

10 ng/ml LIF as well as once after 14 days for the one containing 1 pg. The<br />

SSEA-1 expression remains at a level of above 97% when 10 ng LIF/ml<br />

were used, but is broadly decreased for the negative control. For the<br />

concentration of 1 pg LIF/ml SSEA-1 expression levels of about 90% after<br />

14 days were achievable although this amount of LIF showed no<br />

differences from the negative control in regards of cell viability. From<br />

these results we can state that with regard to the pluripotency maintaining<br />

potential the different LIF did not show detectable differences.<br />

Conclusion: The production and isolation of LIF from E.coli as well as<br />

from CHO cells was successfully established. The bioactivity of both<br />

proteins was demonstrated in various experiment using different cells<br />

and methods of detection. We conclude from our results that LIF from<br />

mammalian cells and LIF from prokaroytes are equally effective.<br />

Acknowledgement: This work was performed within the activities of the<br />

JRG “large scale cultivation” of the DFG cluster of excellence “Rebirth”<br />

(EXC 62/1)<br />

References<br />

1. Tomala M, Lavrentieva A, Moretti P, Rinas U, Kasper C, Stahl F,<br />

Schambach A, Warlich E, Martin U, Cantz T, Scheper T: Preparation of<br />

bioactive soluble human leukemia inhibitory factor from recombinant<br />

Escherichia coli using thioredoxin as fusion partner. Protein Expr Purif<br />

2010, 73(1):51-7.<br />

Page 149 of 181<br />

2. Szabó PE, Hübner K, Schöler H, Mann JR: Allele-specific expression of<br />

imprinted genes in mouse migratory primordial germ cells. Mech Dev<br />

2002, 115:157-160.<br />

P110<br />

Disulphide bond reduction of a therapeutic monoclonal antibody<br />

during cell culture manufacturing operations<br />

Brian Mullan 1* , Bryan Dravis 2 , Amareth Lim 3 , Ambrose Clarke 4 , Susan Janes 3 ,<br />

Pete Lambooy 2 , Don Olson 2 , Tomas O’Riordan 1 , Bruce Ricart 3 ,<br />

Alexander G Tulloch 2<br />

1 Manufacturing Science and Technology, Eli Lilly & Co, Kinsale, Cork, Ireland;<br />

2 Bioprocess R&D, Eli Lilly & Co, Indianapolis, Indiana, USA; 3 Bioproduct<br />

Analytical Chemistry, Eli Lilly & Co, Indianapolis, Indiana, USA; 4 Analytical<br />

Technical Operations, Eli Lilly & Co, Kinsale, Cork, Ireland<br />

E-mail: mullan_brian@lilly.com<br />

BMC Proceedings 2011, 5(Suppl 8):P110<br />

Background: Disulphide bonding is critical to maintaining immunoglobulin<br />

(IgG) tertiary and quaternary structure for therapeutic monoclonal<br />

antibodies (MAb). Both inter- and intra-chain disulphide bonds are formed<br />

intracellularly in the expression host prior to secretion and purification<br />

during MAb production processes. Disulphide bond shuffling has<br />

previously been reported for IgG 2[1,2] and disulphide-mediated armexchange<br />

for IgG 4[3,4], reflecting innate behaviour of these IgG classes.<br />

However, atypical and significant reduction of disulphide bonds has been<br />

recently observed in IgG1[5,6] that present significant issues for<br />

manufacturing of therapeutic MAbs.<br />

During manufacturing of preliminary lots of a recently transferred MAb<br />

manufacturing process (IgG 1), gross disulphide bond reduction following<br />

affinity capture chromatography of clarified production bioreactor<br />

material was observed. Investigations leading to the identification of the<br />

nature of this reduction process, and process steps to mitigate against its<br />

future occurrence, are described here. The MAb was co-developed with<br />

MacroGenics, Rockville, MD.<br />

Methods: Production Bioreactor material for downstream processing was<br />

supplied from a 16 day, fed-batch, GS-CHO culture [7]. The Production<br />

Bioreactor was a single-use (Wave System) with a 100L (full scale) or 10L<br />

(lab model) working volume. Clarified harvest intermediate (CHI) hold<br />

studies were performed in either 560L LevMix units (full scale) or 5L<br />

Braun benchtop bioreactors (lab model). Purification to produce Affinity<br />

Capture chromatography eluted mainstream was performed using a<br />

20cm x 20cm MAbselect Protein A resin (GE Healthcare) column, and an<br />

AKTA Process skid (GE Healthcare).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P110) IgG disulphide bond reduction under various conditions for cell-settled and immediately clarified harvest material. CHI, Clarified<br />

harvest intermediate; DiS, Disulphide; LoC, Lab-on-a-Chip (Agilent).<br />

LC-MS analysis was performed on a Polymer Laboratories PLRP-S HPLC<br />

column and analyzed using an Agilent 1100 HPLC system coupled to<br />

an Applied Biosystems QSTAR XL mass spectrometer, following sample<br />

preparation. CE-SDS analysis was performed using a Beckman Coulter<br />

PA800 capillary electrophoresis instrument fitted with bare-fused silica<br />

capillary and UV detection at 220 nm, following sample preparation.<br />

Microchip CE-SDS analysis was performed using a Lab-on-chip<br />

microanalyser (Agilent). Free thiols were quantified using Ellman’s<br />

reagent. Metabolic analysis was conducted by Metabolon (Durham,<br />

NC).<br />

Results:<br />

Identification of disulphide reduction of IgG during Primary Recovery:<br />

The IgG manufacturing process as transferred from the co-developing<br />

partner included a cell culture settling step following the Production<br />

Bioreactor and prior to Primary Recovery.<br />

Disulphide bond reduction was first detected during initial development<br />

runs by routine Non-reduced (NR) CE-SDS in-process analysis after Affinity<br />

capture chromatography (data not shown). NR-CE-SDS analysis identified<br />

elevated levels of free light chain and half antibody molecules, when<br />

compared to Reference Standard.<br />

Additional analysis, employing microchip-based NR-CE-SDS methods<br />

indicated that the antibody reduction occurred during the primary recovery<br />

cell settling step (results not shown). This was confirmed by LC-MC analysis<br />

(results not shown). Assessment of disulphide bonding pattern and<br />

intactness by LC-MS peptide mapping identified both inter- and intra-chain<br />

disulphide scrambling (results not shown).<br />

Delineation of events leading to IgG reduction: Initial investigations to<br />

understand process behaviour during primary recovery identified that<br />

reducing species, including free thiols (which increase over the course of<br />

the Production Bioreactor, up to 1mM), were present at the end of the<br />

Production Bioreactor (Figure 1a). Dissolved oxygen was also shown to<br />

deplete during the cell settling phase following harvest (data not shown).<br />

From this, an initial working hypothesis was formed that reducing<br />

Page 150 of 181<br />

species, including free thiols, became reactive at low dissolved oxygen<br />

concentrations and led to IgG 1 disulphide bond reduction.<br />

A revised process control strategy was implemented (see below) to<br />

prevent oxygen depletion and maintain dissolved oxygen levels above a<br />

minimum level. This involved including an aerated and agitated hold for<br />

Clarified Harvest Intermediate (CHI) in the process.<br />

Further studies identified that O 2 is critical to maintaining a stable<br />

environment for oxidised (i.e., normally disulphide bonded) IgG 1 in CHI.<br />

When O 2 was present, IgG 1 remained intact under all conditions<br />

evaluated. Only when O 2 was deliberately absent, or stripped away,<br />

would the harvest material or CHI demonstrate potential for reduction<br />

(Figure 1b).<br />

Metabolic behaviour of reducing intermediates: The working hypothesis<br />

was that by maintaining sufficient levels of dissolved oxygen in the CFM, the<br />

thiol species could be reacted out (oxidised) and a stable environment for<br />

oxidised IgG 1 created (Figure 1a). However, the relationship between IgG<br />

reduction and thiol redox state is not first order (Figure 1c), and the rate of<br />

thiol oxidation was found to be dependent on the source of Production<br />

Bioreactor material (i.e., varied with different harvest lots). This indicated the<br />

involvement of an additional component, potentially catalytic, which has not<br />

yet been identified in our studies. Thioredoxins have been identified as such<br />

a catalytic component by others [5,6], and these need to be recycled after<br />

one redox cycle via Thioredoxin Reductase / NADP(H).<br />

Metabolic analysis of cell and media material from Production Bioreactors<br />

indicated high levels of oxidised homocysteine and cysteine (both reactive<br />

redox molecules), which correlated with decreasing levels of folate (B6) and<br />

cobalamine (B12), both of which are involved in recycling homocysteine.<br />

Overall, this analysis identified numerous options for media optimisation to<br />

mitigate against IgG reduction. However, given the success of process<br />

controls (described below), and the late stage of process development (prevalidation)<br />

these media optimisation options were not pursued.<br />

Process controls to mitigate disulphide reduction of IgG: Aprocess<br />

control strategy was implemented including:


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Establishing a minimal dissolved oxygen level in the Production<br />

Bioreactor prior to harvesting<br />

Immediately clarifying the Production Bioreactor material (i.e.,<br />

eliminating the cell settling step)<br />

Holding the CHI in a hold vessel (LevMix container, agitated hold) that<br />

had been partially pre-filled with process air.<br />

Conclusions: Gross disulphide bond reduction was observed during late<br />

stage development of an IgG 1 monoclonal antibody being<br />

commercialised for a therapeutic indication;<br />

Disulphide bond reduction had a second, or higher, order link to low<br />

dissolved oxygen levels in process intermediates, and the involvement of<br />

a catalytic factor was also indicated;<br />

Implementation of an appropriate control strategy (and associated<br />

process analytics) informed by process development has ensured no<br />

recurrence of this issue (for n=15 full scale lots).<br />

References<br />

1. Wypych J, et al: Human IgG2 antibodies display disulfide-mediated<br />

structural isoforms. J Biol Chem 2008, 283:16194-16205.<br />

2. Liu YD, et al: Human IgG2 antibody disulfide rearrangement in vivo.<br />

J Biol Chem 2008, 283:29266-29272.<br />

3. van der Neut Kolfschoten M: Anti-inflammatory activity of human IgG4<br />

antibodies by dynamic Fab arm exchange. Science 2007, 317:1554-1557.<br />

4. Labrijn AF, et al: Therapeutic IgG4 antibodies engage in Fab-arm<br />

exchange with endogenous human IgG4 in vivo. Nature Biotechnology<br />

2009, 27:767-773.<br />

5. Trexler-Schmidt M, et al: Identification and Prevention of Antibody<br />

Disulfide Bond Reduction During Cell Culture Manufacturing.<br />

Biotechnology and Bioengineering 2010, 106:452- 461.<br />

6. Kao Y-H, et al: Mechanism of Antibody Reduction in Cell Culture<br />

Production Processes. Biotechnology and Bioengineering 2010, 107:622-632.<br />

7. Mullan B, et al: Transfer, Implementation and Late Stage Development of<br />

an End-To-End Single-Use Process for Monoclonal Antibody<br />

Manufacture. American Pharmaceutical Review 2011, 58-64.<br />

P111<br />

Preliminary evaluation of microcarrier culture for growth and<br />

monoclonal antibody production of CHO-K1 cells<br />

M Elisa Rodrigues, Pedro Fernandes, A Rita Costa, Mariana Henriques * ,<br />

Joana Azeredo, Rosário Oliveira<br />

IBB-Institute for Biotechnology and Bioengineering, Centre of Biological<br />

Engineering, University of Minho, Braga, Portugal<br />

E-mail: mcrh@deb.uminho.pt<br />

BMC Proceedings 2011, 5(Suppl 8):P111<br />

Background: The large-scale production of biopharmaceuticals commonly<br />

relies on the use of suspension cell cultures, since they provide higher yields<br />

than adherent cultures. However, most mammalian cells grow in adherent<br />

mode and therefore need to go through a process of adaptation to<br />

suspended growth, which is not always simple or feasible. In this context,<br />

microcarrier cultures have introduced new possibilities, allowing the<br />

practical culture of anchorage-dependent cells, in suspension systems,<br />

achieving high yields. In these systems, cells grow as monolayers on the<br />

surface of small spheres – the microcarriers, which are usually kept in<br />

suspension in the culture medium by gentle rocking. The aim of the present<br />

study was to evaluate, compare and optimize the use of microcarrier culture<br />

for the growth and monoclonal antibody (mAb) production of CHO-K1 cells.<br />

Material and methods: Two types of microcarriers were assessed and<br />

compared in this study: macroporous and microporous. For this, cultures of<br />

mAb-producing CHO-K1 cells were performed in vented conical tubes, at<br />

37 °C and 5% CO 2. CHO-K1 culture assessment was divided in two phases:<br />

the initial cell adhesion phase; and the cell proliferation phase. A set of<br />

different conditions was tested, namely: initial cell concentration (2x10 5<br />

cells/ml and 4x10 5 cells/ml), microcarrier concentration (1 g/L for<br />

macroporous and 3 g/L for microporous), type of rocking during the initial<br />

phase of adhesion (continuous and pulse) and during the cell proliferation<br />

phase (continuous). Medium was renewed on a daily basis and the<br />

concentration and viability of cells adhered to the microcarriers were<br />

periodically assessed (hourly for the adhesion phase, and daily after that).<br />

Furthermore, samples were taken for antibody quantification by enzymelinked<br />

immunosorbent assay (ELISA).<br />

Page 151 of 181<br />

Results: Concerning the phase of initial cell adhesion to the microcarriers,<br />

it was observed that cell adhesion to the microporous microcarriers is<br />

favored by the use of a higher initial cell concentration (4x10 5 cells/ml)<br />

with both pulse and particularly continuous rocking methodologies. On<br />

the other hand, cell adhesion to the macroporous microcarriers is favored<br />

by a higher initial cell concentration, but only with continuous rocking.<br />

For a lower initial cell concentration, a pulse rocking methodology is<br />

recommended. For both microcarriers, the majority of cell adhesion<br />

occurs within the first 3 hours.<br />

Regarding the cell proliferation phase, the results showed that it is affected<br />

by the inoculum concentration only for the microporous microcarriers, with<br />

4x10 5 cells/ml providing the best cell proliferation. Comparing the two types<br />

of microcarriers in terms of cell growth, it was observed that the<br />

microporous provided higher cell proliferation than the macroporous.<br />

Additionally, the microporous microcarriers demonstrated a higher durability<br />

than the macroporous, which starts to disintegrate after two weeks.<br />

Concerning the results of mAb production, it was observed that in<br />

microporous cultures it is favored by the use of a pulse rocking<br />

methodology in the initial phase of adhesion, for both inoculum<br />

concentrations evaluated. This was also observed in macroporous cultures<br />

but only for the lowest concentration of cells. With 4x10 5 cells/ml the use<br />

of a continuous rocking methodology proved to be advantageous.<br />

Indeed, the highest level of production and productivity was achieved in<br />

these conditions – 4x10 5 cells/ml and continuous rocking. Furthermore,<br />

the results show that the cells have higher productivities when cultured<br />

in macroporous microcarriers than inmicroporous,inspiteofhaving<br />

better levels of proliferation in the last one. Indeed, with fewer cells, the<br />

macroporous carrier was able to provide levels of total mAb production<br />

similar and even greater than the microporous.<br />

Conclusions: This study demonstrated that microcarrier cultures are a<br />

viable alternative to suspended cultures for the growth and antibody<br />

production of CHO-K1 cells. For this purpose, the use of higher inoculum<br />

concentrations during the initial phase of cell adhesion is particularly<br />

favorable if continuous rocking is used.<br />

The comparison of the two different types of microcarriers assessed<br />

indicated that, in general, higher levels of cell adhesion and proliferation are<br />

obtained with microporous microcarriers, while higher mAb productivity<br />

and total production are achieved with the macroporous. Therefore, the<br />

microporous microcarriers assessed is recommended for purposes of cell<br />

growth while the macroporous is indicated for purposes of production.<br />

Among the culture conditions tested, the most favorable for the purpose of<br />

mAb production is the use of the macroporous microcarriers cultured at<br />

4x10 5 cells/ml under continuous rocking.<br />

P112<br />

Strategies for adaptation of mAb-producing CHO cells to serum-free<br />

medium<br />

A Rita Costa, M Elisa Rodrigues, Mariana Henriques * , Rosário Oliveira,<br />

Joana Azeredo<br />

IBB-Institute for Biotechnology and Bioengineering, Centre of Biological<br />

Engineering, University of Minho, Braga, Portugal<br />

E-mail: mcrh@deb.uminho.pt<br />

BMC Proceedings 2011, 5(Suppl 8):P112<br />

Background: The large-scale production of biopharmaceuticals, such as<br />

monoclonal antibodies (mAbs), commonly requires the use of serum-free<br />

medium, for both safety and economical reasons. However, because serum<br />

is such an essential supplement of the growth medium of most mammalian<br />

cells, its removal demands a very time-consuming process of cell adaptation.<br />

In this process, cells are usually subjected to a gradual, step-wise, decrease<br />

of serum concentration in the medium. With the purpose of alleviating cell<br />

adaptation, other medium supplements such as insulin and trace elements<br />

can be used, either isolated or in combination. Thus, the aim of this study<br />

was to assess strategies for the adaptation of CHO cells to serum-free media,<br />

using different supplement combinations, as well as to identify the most<br />

critical steps of the process.<br />

Materials and methods: The study was divided in two experiments. In the<br />

first one, cultures of mAb-producing CHO-K1 cells were initiated in 24 well<br />

plates, using Dulbecco’s Modified Eagle Medium (DMEM) supplemented<br />

with 10% serum. The effect of five combinations of supplements that could


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

support cells during adaptation was tested. These supplements included<br />

insulin and eight different trace elements. A methodology of gradual<br />

adaptation was followed, consisting of sequential steps of serum reduction.<br />

At the level of 0.625% serum the medium was gradually switched from<br />

DMEM to the chemically-defined serum-free EX-CELL CHO DHFR - medium.<br />

The second experiment of this study was performed in order to overcome<br />

the problems identified in the first assay. The adaptation methodology<br />

was similar, with the following changes: cell cultures were initiated in 25<br />

cm 2 T-flasks, and only three of the initial five combinations of<br />

supplements were tested.<br />

Results: In the first experiment, cells growing in medium supplemented<br />

with the two combinations containing a trace element in common died at<br />

2.5% serum, while for the remaining combinations cell death occurred at a<br />

later stage, 0.625% serum. Cell death was attributed to problems with the<br />

procedure of adaptation used in the first experiment, which were identified<br />

and corrected in the second experiment. The problems found and the<br />

procedure modifications implemented included the use of higher initial cell<br />

concentrations to allow the survival of an increased number of cells during<br />

the process; avoiding procedures that can be harsh to the cells, such as<br />

centrifugation and the use of enzymes (i.e. trypsin) due to a higher<br />

sensitivity of cells during adaptation; and allowing enough time in each step<br />

of the process for a complete cell adaptation.<br />

After these modifications, in the second experiment, it was possible to<br />

observe that cells required a long time to adapt to each level of serum<br />

concentration, particularly below 0.625%. At the level of 0.31% serum, cells<br />

start to detach, and become fully detached at 0.15%, growing in suspension<br />

from this point on. The 0.31% of serum was the most critical step of the<br />

process, demanding more time for cell adaptation and causing the death of<br />

cells growing in two of the combinations assayed. Indeed, only cells<br />

growing in medium supplemented with one of the combinations were able<br />

to survive the whole process.<br />

It should also be noted that adaptation of cells to EX-CELL is easily achieved<br />

as long as some serum supplementation is maintained.<br />

Conclusions: This study demonstrated that the process of adaptation to<br />

serum-free medium is very challenging to the cells. They become extremely<br />

sensitive to common cell culture procedures, such as centrifugation or the<br />

use of enzymes, and consequently extra care should be taken when<br />

developing the adaptation procedure. Furthermore, it was shown that cell<br />

adaptation to serum-free conditions is affected by the medium supplements<br />

used, as well as the time given to each step of the process.<br />

P113<br />

Impact of different influenza cultivation conditions on HA<br />

N-Glycosylation<br />

Jana V Roedig 1 , Erdmann Rapp 1* , Yvonne Genzel 1 , Udo Reichl 1,2<br />

1 Max Planck Institute for Dynamics of Complex Technical Systems,<br />

Sandtorstraße 1, Magdeburg, Germany; 2 Otto-von-Guericke-University, Chair<br />

of Bioprocess Engineering, Magdeburg, Germany<br />

E-mail: rapp@mpi-magdeburg.mpg.de<br />

BMC Proceedings 2011, 5(Suppl 8):P113<br />

Background: Influenza virus is a highly contagious human and animal<br />

pathogen causing infections of the respiratory track. Prevention such as<br />

high standard hygiene and vaccination still represent the best measures<br />

for protection. Beside the traditional egg-based influenza vaccine<br />

production, numerous cell culture-based processes are currently being<br />

established. Due to its ability to induce strong and protective immune<br />

responses, the highly abundant glycoprotein hemagglutinin (HA)<br />

represents the major component in influenza vaccines. Since variations<br />

in N-glycosylation of glycoproteins such as HA can alter quality<br />

characteristics of antigens, the impact of cell lines and process<br />

parameters for vaccine manufacturing needs to be addressed. This<br />

study investigates the impact of virus adaptation and different harvest<br />

time points on HA N-glycosylation. Therefore, the HA of influenza A<br />

virus Uruguay/716/2007 (H3N2, high growth reassortant), in the<br />

following referred to as IVA-Uruguay, was purified and N-glycans<br />

analyzed by capillary gel electrophoresis with laser-induced fluorescence<br />

(CGE-LIF).<br />

Materials and methods:<br />

Cell culture and virus production: IVA-Uruguay (H3N2, #07/360, NIBSC,<br />

South Mimms, UK) was produced in either adherently growing MDCK<br />

Page 152 of 181<br />

(No. 84121903) or Vero (No. 88020401) cells purchased from ECACC<br />

(Salisbury, UK). For cell growth GMEM (Invitrogen, #22100-093, Darmstadt,<br />

Germany) was supplemented with 5.5 g/L glucose (Roth, #X997.3,<br />

Karlsruhe, Germany), 2 g/L peptone (IDG, #MC33, Lancashire, UK), 10%<br />

FCS (Invitrogen, #10270-106) and 4 mg/mL NaHCO3 (Roth, #6885.3).<br />

Infections were performed in the same medium without addition of FCS<br />

but supplemented with trypsin (Invitrogen, #27250-018) at a final<br />

concentration of 5 U/mL. Virus was quantified by a hemagglutination<br />

assay according to Kalbfuss et al. [1] and is expressed in HAU (log HA/<br />

100 µL).<br />

HA N-glycosylation pattern analysis: Virus was harvested and processed<br />

for HA N-glycosylation pattern analysis according to Schwarzer et al. [2]<br />

applying an optimized work-flow [3] and data evaluation [4]. Finally, the<br />

samples were separated by CGE-LIF using an ABI PRISM 3100-Avant<br />

genetic analyzer (Applied Biosystems, Foster City, California, USA). For data<br />

processing and evaluation the x-axis of capillary electropherograms was<br />

normalized using an internal standard, resulting in N-glycosylation patterns,<br />

in which each peak corresponds to at least one distinct N-glycan structure.<br />

This allowed a direct qualitative comparison regarding N-glycan structure<br />

presence in different samples. For quantitative comparison, the relative<br />

peak height (RPH: the ratio of peak height to the total height of all peaks)<br />

was determined for each peak and sample. Low abundant peaks were<br />

defined with RPH < 5%.<br />

Results: MDCK cell-derived virus seed, exhibiting 2.6 HAU at 24 hours post<br />

infection (hpi; data not shown), was used to infect five consecutive<br />

passages of Vero cells. Adaptation of the virus to Vero cells resulted in<br />

increased virus yields within shorter time frames in the new host system:<br />

in the first passage of Vero cells 2.1 HAU were obtained at 96 hpi, whereas<br />

in the fifth passage a titer of 2.7 HAU at 72 hpi was reached (data not<br />

shown). The HA N-glycosylation pattern of the MDCK cell-derived IVA-<br />

Uruguay seed exhibited 25 different characteristic peaks in the range of<br />

160 bp to 400 bp. Of these a total number of 11 peaks representing large<br />

glycans (275 bp - 400 bp; #12, 13, 18 - 22, 24, 26 - 28) were unique to<br />

MDCK cell-derived virus (figure 1A). In contrast, the HA N-glycosylation<br />

pattern changed significantly with the first passage in Vero cells. Here, 15<br />

different peaks between 150 bp and 380 bp characterize the Vero cellspecific<br />

HA N-glycosylation pattern. Four peaks (#3, 5, 23 and 25) were<br />

unique to Vero cell-derived virus (figure 1A). In comparison to MDCK cellderived<br />

HA, the Vero cell-derived antigen showed a tendency towards<br />

smaller glycan structures. The relative abundance of each peak over all<br />

Vero passages only varied marginally with standard deviations (SD) ≤ 2.1%<br />

(table 1). The increase in virus titers within shorter time frames suggests<br />

increased viral fitness, during adaptation from MDCK to Vero cells. An<br />

impact of HA N-glycosylation on properties of the virus, i.e. virus<br />

replication, has already been descried [8-12]. Interestingly, the glycan<br />

pattern stabilized soon after the first passage in Vero cells. This clearly<br />

indicates that further increase in HA titer did not depend on changes in<br />

the HA N-glycosylation pattern.<br />

The impact of harvest time on the HA N-glycosylation pattern of MDCK<br />

cell-derived IVA-Uruguay is shown in figure 1B. Virus harvested at either<br />

24 hpi or 72 hpi exhibited the 25 different MDCK cell-specific peaks<br />

between 160 bp and 400 bp. At 72 hpi one additional, but very low<br />

abundant peak was detected (numbered 23). Overall, differences in<br />

relative structure abundance were rather small with a maximal difference<br />

of 1.7% RPH (table 1). This indicates that HA of virus particles released in<br />

the supernatant is rather stable over the time window relevant for<br />

influenza virus production [5,6].<br />

However, there are minor variations in RPH from passage to passage<br />

during adaptation and between harvesting time points. Possible<br />

explanations are varying ratios either of completely/incompletely<br />

processed or of intact/degraded N-glycan structures or a combination of<br />

both. In 2009, Schwarzer et al. [7] characterized the MDCK cell-derived HA<br />

N-glycosylation pattern of a H3N2 influenza virus subtype as a mixture of<br />

complex N-glycan structures with terminal a- andb-galactose and high<br />

mannose type structures. In contrast, the Vero cell-derived HA was<br />

characterized by complex N-glycans with exclusively terminal b-galactose<br />

and structures of the high mannose type. For final evaluation of the<br />

results presented here, determination of the N-glycan structure of all<br />

peaks would be required.<br />

Conclusion: In this study, the impact of adaptation and harvesting time<br />

point on HA N-glycosylation of IVA-Uruguay was investigated. So far, it is<br />

not clear whether differences in the HA N-glycosylation have an impact


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Page 153 of 181<br />

Figure 1(abstract P113) HA N-glycosylation patterns of Influenza A virus Uruguay/716/2007 (H3N2) – high growth reassortant. Relative<br />

fluorescence units (RFU) are plotted over the migration time (t mig) in base pairs (bp). (A) MDCK cell-derived virus (seed) was consecutively passaged in<br />

Vero cells (1 st passage to 5 th passage). Peaks annotated in blue are present in MDCK as well as Vero cell-derived HA; peaks annotated in black are MDCK<br />

cell-specific, and red annotation indicates Vero cell-specific peaks. (B) MDCK cell-derived virus harvested either 24 or 72 hours post infection (hpi). MDCK<br />

cell-specific peaks are annotated (1, 2, 4, 6 – 24, 26-28).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P113) Overview of relative peak heights (RPH) of 28 HA-glycan peaks during virus adaptation from<br />

MDCK to Vero cells, and of two harvest time points in a cultivation with MDCK cells. For virus adaptation, MDCK cellderived<br />

virus seed was consecutively passaged in Vero cells (pass. 1 to pass. 5). Two harvest time points, 24 and 72 hours<br />

post infection (hpi), compared by absolute values of RPH and percentage difference |ΔRPH|. Maximal SD and maximal |ΔRPH|<br />

are highlighted in bold<br />

Peak Virus Adaptation Time Course<br />

seed pass. 1 pass. 2 pass. 3 pass. 4 pass. 5 pass. 1 to 5 24 hpi 72 hpi |ΔRPH|<br />

No. Relative Peak Height (RPH) [%] SDRPH* [%] RPH [%] [%]<br />

1 7,8 0,9 1,7 0,9 2,2 4,8 1,6 3,9 4,5 0,6<br />

2 3,9 5,7 4,6 5,1 6,1 4,3 0,7 2,4 2,8 0,4<br />

3 0,0 2,2 1,9 3,4 1,4 1,4 0,8 0 0 0<br />

4 8,1 6,6 5,9 6,1 6,8 6,8 0,4 5,3 5,2 0,1<br />

5 0,0 4,0 2,7 2,5 2,5 1,9 0,8 0 0 0<br />

6 1,4 2,5 1,9 1,5 1,5 1,7 0,4 2,1 1,7 0,4<br />

7 6,2 4,4 4,8 4,1 4,7 4,8 0,3 4,0 3,6 0,4<br />

8 6,5 10,0 9,6 8,1 7,4 7,6 1,2 5,3 5,2 0,1<br />

9 1,7 4,3 3,4 3,4 4,1 5,6 0,9 1,1 1,1 0<br />

10 13,7 25,8 23,5 27,2 28,6 24,3 2,1 17,6 16,3 1,3<br />

11 6,8 11,8 12,3 12,2 11,7 9,5 1,2 5,5 4,9 0,6<br />

12 6,2 0 0 0 0 0 0 2,5 2,5 0<br />

13 2,0 0 0 0 0 0 0 4,6 4,4 0,2<br />

14 3,1 0,6 1,1 1,9 1,3 1,5 0,5 3,7 3,6 0,1<br />

15 6,3 4,1 4,1 4,4 3,8 3,5 0,3 5,0 4,7 0,3<br />

16 2,2 7,7 7,4 7,4 7,8 8,9 0,6 5,9 6,0 0,1<br />

17 1,6 3,3 4,4 3,3 3,1 4,3 0,6 2,3 2,8 0,5<br />

18 1,3 0 0 0 0 0 0 1,9 2,3 0,4<br />

19 4,5 0 0 0 0 0 0 3,6 4,1 0,5<br />

20 2,0 0 0 0 0 0 0 8,0 7,0 1,0<br />

21 2,6 0 0 0 0 0 0 1,7 2,2 0,5<br />

22 3,3 0 0 0 0 0 0 3,9 3,8 0,1<br />

23 0,0 4,7 7,9 6,2 5,0 6,4 1,3 0 1,7 1,7<br />

24 1,9 0 0 0 0 0 0 1,6 1,7 0,1<br />

25 0,0 1,5 3,0 2,3 1,9 2,6 0,6 0 0 0<br />

26 2,4 0 0 0 0 0 0 2,2 2,1 0,1<br />

27 1,1 0 0 0 0 0 0 2,3 2,3 0<br />

28 3,1 0 0 0 0 0 0 3,7 3,4 0,3<br />

* standard deviation (SDRPH) of all characteristic peaks in MDCK and Vero cells during virus adaptation.<br />

on immunogenicity or other properties of influenza vaccines. Other<br />

factors, e.g. differences in cell culture media, cell density, etc. may also<br />

contribute to variations in HA N-glycosylation. Nevertheless, monitoring<br />

N-glycosylation patterns during vaccine production processes allows not<br />

only to evaluate antigen quality and the impact of process modifications<br />

on lot-to-lot consistency but also to critically assess consequences of<br />

unwanted process variations or process failure.<br />

References<br />

1. Kalbfuss B, Knochlein A, Krober T, Reichl U: Monitoring influenza virus<br />

content in vaccine production: precise assays for the quantitation of<br />

hemagglutination and neuraminidase activity. Biologicals 2008,<br />

36(3):145-161.<br />

2. Schwarzer J, Rapp E, Reichl U: N-glycan analysis by CGE-LIF: profiling<br />

influenza A virus hemagglutinin N-glycosylation during vaccine<br />

production. Electrophoresis 2008, 29(20):4203-4214.<br />

3. Rödig J, Rapp E, Hennig R, Schwarzer J, Reichl U: Optimized CGE-LIF-Based<br />

Glycan Analysis for High-Throughput Applications. Proceedings of the 21st<br />

Annual Meeting of the European Society for Animal Cell Technology (ESACT)<br />

Dublin, Ireland: Springer Science+Business Media B.V. 2009 in press.<br />

4. Ruhaak LR, Hennig R, Huhn C, Borowiak M, Dolhain RJ, Deelder AM, Rapp E,<br />

Wuhrer M: Optimized workflow for preparation of APTS-labeled N-<br />

Page 154 of 181<br />

glycans allowing high-throughput analysis of human plasma glycomes<br />

using 48-channel multiplexed CGE-LIF. J Proteome Res 2010,<br />

9(12):6655-6664.<br />

5. Tree JA, Richardson C, Fooks AR, Clegg JC, Looby D: Comparison of largescale<br />

mammalian cell culture systems with egg culture for the<br />

production of influenza virus A vaccine strains. Vaccine 2001, 19(25-<br />

26):3444-3450.<br />

6. Aggarwal K, Jing F, Maranga L, Liu J: Bioprocess optimization for cell<br />

culture based influenza vaccine production. Vaccine 2011,<br />

29(17):3320-3328.<br />

7. Schwarzer J, Rapp E, Hennig R, Genzel Y, Jordan I, Sandig V, Reichl U:<br />

Glycan analysis in cell culture-based influenza vaccine production:<br />

influence of host cell line and virus strain on the glycosylation pattern<br />

of viral hemagglutinin. Vaccine 2009, 27(32):4325-4336.<br />

8. Tsuchiya E, Sugawara K, Hongo S, Matsuzaki Y, Muraki Y, Li ZN, Nakamura K:<br />

Effect of addition of new oligosaccharide chains to the globular head of<br />

influenza A/H2N2 virus haemagglutinin on the intracellular transport and<br />

biological activities of the molecule. J Gen Virol 2002, 83(Pt 5):1137-1146.<br />

9. Deshpande KL, Fried VA, Ando M, Webster RG: Glycosylation affects<br />

cleavage of an H5N2 influenza virus hemagglutinin and regulates<br />

virulence. Proc Natl Acad Sci U S A 1987, 84(1):36-40.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

10. Wang CC, Chen JR, Tseng YC, Hsu CH, Hung YF, Chen SW, Chen CM,<br />

Khoo KH, Cheng TJ, Cheng YS, et al: Glycans on influenza hemagglutinin<br />

affect receptor binding and immune response. Proc Natl Acad Sci U S A<br />

2009, 106(43):18137-18142.<br />

11. Klenk HD, Wagner R, Heuer D, Wolff T: Importance of hemagglutinin<br />

glycosylation for the biological functions of influenza virus. Virus Res<br />

2002, 82(1-2):73-75.<br />

12. Wagner R, Heuer D, Wolff T, Herwig A, Klenk HD: N-Glycans attached to<br />

the stem domain of haemagglutinin efficiently regulate influenza A<br />

virus replication. J Gen Virol 2002, 83(Pt 3):601-609.<br />

P114<br />

Effect of iron sources on the glycosylation macroheterogeneity of human<br />

recombinant IFN-g produced by CHO cells during batch processes<br />

Marie-Françoise Clincke 1,2,3 , Emmanuel Guedon 1* , Frances T Yen 2 ,<br />

Virginie Ogier 3 , Jean-Louis Goergen 1<br />

1 Laboratoire Réactions et Génie des Procédés UPR-CNRS 3349, ENSAIA-INPL,<br />

Nancy-Université, 54505 Vandoeuvre-lès-Nancy, France; 2 Lipidomix (EA4422),<br />

ENSAIA-INPL, Nancy-Université, 54505 Vandoeuvre-lès-Nancy, France; 3 Genclis<br />

SAS, 54505 Vandoeuvre-lès-Nancy, France<br />

E-mail: Emmanuel.Guedon@ensaia.inpl-nancy.fr<br />

BMC Proceedings 2011, 5(Suppl 8):P114<br />

Background: In the biopharmaceutical industry, the control of<br />

glycosylation to satisfy the quality consistency of recombinant proteins<br />

produced during a process has become an important issue. Indeed, the<br />

glycosylation pattern of recombinant proteins could be influenced by<br />

different factors including the cell line used, environmental factors such<br />

as oxygenation, temperature, shear stresses, extracellular pH... and the<br />

availability of nutrients.<br />

As previously reported [1,2], the BDM medium is able to support a<br />

better CHO cell growth and a higher IFN-g production compared to<br />

RPMI medium supplemented with serum. In addition, when BDM<br />

medium is used, CHO cells are capable to maintain a high percentage<br />

of doubly-glycosylated glycoforms of recombinant IFN-g produced<br />

during the whole process. Conversely, mono-glycosylated and nonglycosylated<br />

IFN-g forms increased during batch cultures performed<br />

with RPMI serum medium.<br />

Iron is an important nutrient and is reported to support essential<br />

functions in cells. In this study, the impact of different iron sources on<br />

the CHO cell growth, as well as on the production and the glycosylation<br />

of a human recombinant IFN-g were investigated.<br />

Materials and methods: CHO cell cultures producing human recombinant<br />

IFN-g (CHO 320: dhfr+, a2.6 ST-) were performed in Erlenmeyer flasks (37°C,<br />

pH 7.2, 70 rpm) and in two different media, namely RPMI and BDM [3].<br />

Whereas RPMI is a classical medium containing serum (5%), BDM is<br />

Page 155 of 181<br />

a chemically defined medium without any proteins or serum addition.<br />

BDM was supplemented with 0.1% pluronic F-68, 750 µM ethanolamine<br />

and 500 µM iron citrate.<br />

IFN-g was quantified using an Elisa test. Glycosylation macroheterogeneity<br />

of IFN-g was characterized by Western Blotting (Amersham Biosciences)<br />

and each glycoform was quantified by densitometry as previously<br />

described [2].<br />

Results: CHO cell growth, IFN-g production and quality:<br />

CHO cell cultures producing human recombinant IFN-g were cultivated in<br />

Erlenmeyerflasks,inbothRPMIsupplementedwith5%SVFandBDM<br />

media. Kinetics performed in BDM medium resulted in a higher maximal<br />

viable cell density and IFN-g production compared to RPMI medium (data<br />

not shown). In fact, the higher IFN-g production by CHO cells observed in<br />

BDM medium was mostly due to a higher cell density, since the specific<br />

rate of IFN-g production ( qIFN-g) was slighly higher in the BDM medium<br />

than in the RPMI serum medium.<br />

In both media, three major molecular weight variants (2N, 1N, 0N) were<br />

detected during the process with a majority of IFN-g doubly-glycosylated<br />

(2N) whatever the medium used (figure 1).<br />

However, the quality of IFN-g remained constant during the CHO cell<br />

cultures performed in BDM medium, contrasting with the increase in the<br />

proportion of non-glycosylated (0N) IFN-g to the detriment of the doublyglycosylated<br />

form (2N), during the time course of the culture of CHO cells<br />

performed in RPMI serum medium.<br />

Addition of iron citrate in RPMI serum strongly affects the cell<br />

growth, the production and the quality of IFN-g:<br />

Supplementation of RPMI serum medium with iron citrate had a strong<br />

effect on kinetics of CHO cells producing IFN-g. Indeed, a higher maximal<br />

viable CHO cell density as well as a high IFN-g specific production rate<br />

were measured, compared to kinetics of CHO cells performed in RPMI<br />

serum medium without any supplementation (data not shown). In<br />

addition, supplementation of RPMI with iron citrate resulted in a constant<br />

glycosylation pattern of IFN-g whereas an increase of non-glycosylated<br />

IFN-g to the detriment of the doubly glycosylated form was observed<br />

when CHO cells are were cultivated in RPMI serum (Figure 1).<br />

Addition of other bioavailable iron sources such as ammoniacal ferric citrate<br />

or selenium iron citrate, in RPMI serum medium resulted in the same<br />

phenomenon. However, when RPMI serum medium was supplemented<br />

with ferric-EDTA, a negative effect on the production of IFN-g was observed<br />

(0.03 mg/10 8 cellules vs. 0.07-0.08 mg/10 8 cellules using iron citrate,<br />

ammoniacal ferric citrate or iron citrate complexed to selenium) despite the<br />

IFN-g macroglycoforms was maintained constant in this condition (data not<br />

shown).<br />

Conclusions: Addition of iron citrate to RPMI serum medium improved<br />

cellgrowth,aswellasIFN-g production. Furthermore, the glycosylation<br />

pattern of IFN-g remained constant when iron citrate was added in the<br />

medium.<br />

Figure 1(abstract P114) Proportion of IFN-g macroglycoforms produced by CHO cells cultivated in various media; RPMI serum, BDM and RPMI serum<br />

supplemented with iron citrate.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Addition of ammoniacal ferric citrate, iron citrate complexed to selenium<br />

or ferric-EDTA to RPMI serum medium also allowed to maintain a<br />

constant macroglycosylation pattern of IFN-g produced during batch<br />

cultures.<br />

However, using ferric-EDTA in supplement of RPMI serum, the specific<br />

production rate of IFN-g was lower compared to values obtained when<br />

other iron sources as named above were used.<br />

Thus, the addition of a bioavailable iron source to culture media could<br />

improve the physiological cell properties as well as the quality of a<br />

recombinant protein expressed.<br />

Acknowledgements<br />

This work was financed by the Agence Nationale pour la Recherche<br />

Technique (ANRT) and Genclis SAS (Vandoeuvre-lès-Nancy, France).<br />

References<br />

1. Mols J, Burteau CC, Verhoeye FR, Ballez JS, Agathos SN, Schneider Y-J:<br />

Fortification of a protein-free cell culture medium with plant peptones<br />

improves cultivation and productivity of an interferon-g producing CHO<br />

cell line. In Vitro Cell Dev Biol Anim 2003, 39:291-296.<br />

2. Kochanowski N, Blanchard F, Cacan R, Chirat F, Guedon E, Marc A,<br />

Goergen J-L: Influence of intracellular nucleotide and nucleotide sugar<br />

contents on recombinant interferon-g glycosylation during batch and<br />

fed-batch cultures of CHO cells. Biotechnol Bioeng 2008, 100:721-733.<br />

3. Schneider Y-J: Optimization of hybridoma cell growth and monoclonal<br />

antibody secretion in chemically defined serum- and protein-free<br />

medium. J Immunol Methods 1989, 116:67-77.<br />

P115<br />

Characterization of metalloprotease and serine protease activities in<br />

batch CHO cell cultures: control of human recombinant IFN-g<br />

proteolysis by addition of iron citrate<br />

Marie-Françoise Clincke 1,2,3 , Emmanuel Guedon 1* , Frances T Yen 2 ,<br />

Virginie Ogier 3 , Jean-Louis Goergen 1<br />

1 Laboratoire Réactions et Génie des Procédés UPR-CNRS 3349, ENSAIA-INPL,<br />

Nancy-Université, 54505 Vandoeuvre-lès-Nancy, France; 2 Lipidomix (EA4422),<br />

ENSAIA-INPL, Nancy-Université, 54505 Vandoeuvre-lès-Nancy, France;<br />

3 Genclis SAS, 54505 Vandoeuvre-lès-Nancy, France<br />

E-mail: Emmanuel.Guedon@ensaia.inpl-nancy.fr<br />

BMC Proceedings 2011, 5(Suppl 8):P115<br />

Background: During the production of any recombinant proteins, an<br />

evaluation of the product quality is crucial. Proteolytic events may<br />

occur during the process and could influence the product quality.<br />

Indeed, proteolysis is an unpredictable process and relatively little is<br />

know regarding the proteolytic enzymes produced and released by<br />

mammalian cells. In fact, proteases originating from the host cell line<br />

cannot be avoided in cell culture. Due to regulatory and safety<br />

prospects, the addition of serum, fetuin or albumin that usually limit<br />

protease activities, is not desirable. Thus, in serum-free cultures of<br />

mammalian cells, control of protease activity constitutes a major<br />

challenge.<br />

In the present work, the presence of proteases and their effect on quality<br />

of IFN-g produced by a recombinant CHO cell line cultivated in a stirredtank<br />

bioreactor were studied. Whereas the quality of IFN-g remained<br />

constant during the CHO cell cultures performed in BDM medium, IFN-g<br />

proteolysis was observed when cultures were carried out in RPMI<br />

medium with serum [1,2].<br />

Materials and methods: IFN-g producing CHO cell lines (CHO 320: dhfr + ,<br />

a2,6 ST - ) were grown in RPMI supplemented with 5% serum and in BDM<br />

medium [3]. Whereas RPMI is a classical medium containing serum, BDM<br />

is a chemically defined medium without any proteins or serum addition,<br />

but supplemented with 0.1% pluronic F-68, 750 µM ethanolamine and<br />

500 µM iron citrate.<br />

Batch cultures were performed in stirred-tank bioreactor (Inceltech, SGI).<br />

Dissolved oxygen concentration was set at 50% of air saturation.<br />

Agitation rate used was 50 rpm; pH and temperature were set at 7.2 and<br />

37°C respectively.<br />

Glycosylation macroheterogeneity of IFN-g was characterized by Western<br />

Blot (Amersham Biosciences).<br />

Gelatinase and caseinase activities were performed using zymography.<br />

Cell-free culture supernatants were concentrated 2-fold on a 10-kDa<br />

Page 156 of 181<br />

cutoff filter. Then, the concentrated samples were instantly mixed 3:1<br />

with nonreducing electrophoresis sample buffer (4.8 mL H 2O; 1.2 mL Tris-<br />

HCl0.5MpH6.8;2mLSDS10%;1mLglycerol;0.5mLbromophenol<br />

blue) and loaded on the zymogram gels. The SDS-PAGE gels (10%<br />

acrylamide) contained either 0.05% caseine or 0.1% gelatin. The gels were<br />

runat25mAfor1h.ToremoveSDS,thegelsweresoakedtwicefor30<br />

min in 2% Triton X-100 on a shaker, then washed in distilled H 2O<br />

followed by an 24h incubation in developing buffer (0.5 M Tris, pH 7.4, 1<br />

µM Zn 2+ and 5 mM Ca 2+ ). Inhibitor supplementations were also<br />

performed using EDTA a , PMSF b and Complete inhibitor Cocktail c .<br />

Visualization of protease activity was carried out by incubation for around<br />

3h in a Coomassie blue solution.<br />

a EDTA (ethylenediaminetetraacetic acid) = inhibitor of metalloprotease<br />

activities<br />

b Complete, Mini, EDTA-free (Roche) = serine and cysteine proteases<br />

inhibitor cocktail<br />

c PMSF (phenylmethylsulfonyl fluoride) = inhibitor of serine protease<br />

activities<br />

Results: CHO cell cultures producing human recombinant IFN-g were<br />

cultivated in stirred-tank bioreactor in both RPMI supplemented with 5%<br />

serum and BDM media. In both media, three major molecular weight<br />

variants (2N, 1N, 0N) were detected during the process with a majority of<br />

IFN-g doubly-glycosylated (2N) whatever the medium used. However,<br />

using the RPMI medium with serum, IFN-g proteolysis was observed<br />

during the whole culture (Figure 1).<br />

To determine the protease activities during CHO cell cultures performed<br />

in both media, zymogram gels containing either gelatin or casein were<br />

performed. Among the 5 caseinase activities detected when CHO cell<br />

cultures were performed with serum (Table 1), only the protease activities<br />

present all over the process could be potentially involved in the IFN-g<br />

proteolysis (220, 90 and 85 kDa). In addition, 2 gelatinase activities were<br />

detected during the whole process (90 and 65 kDa). When CHO cell<br />

cultures were carried out using protein-free BDM medium, 2 caseinase<br />

activities (90 and 85 kDa) and 1 gelatinase activity (90 kDa) were<br />

detected. To determine the type of protease activities present, different<br />

inhibitors were used in zymogram gels containing either casein or<br />

gelatin. EDTA inhibited all the gelatinase activities, identifying these<br />

enzymes as metalloproteases, whereas PMSF and Complete inhibitor<br />

Cocktail inhibited all the caseinase activities, classifying these enzymes as<br />

serine proteases (data not shown).<br />

Compositions of both BDM and RPMI with serum were compared and<br />

3 components which are present in BDM but completely absent in<br />

RPMI were identified. These 3 components are pluronic F-68 (PF-68),<br />

iron citrate and ethanolamine. Interestingly, addition of iron citrate in the<br />

developing buffer of zymogram gels allowed to inhibit the<br />

metalloprotease activities, most likely by blocking the Zinc atom in the<br />

catalytic domain (data not shown). CHO cell cultures were then<br />

performed in RPMI serum supplemented with iron citrate. Addition of<br />

iron citrate to RPMI serum allowed to minimized IFN-g proteolysis (Figure<br />

1). Furthermore, when CHO cell cultures were performed in BDM medium<br />

without iron citrate during the first 30 hours of the culture, IFN-g<br />

proteolysis was detected (data not shown). Therefore, IFN-g proteolysis<br />

has at least one cellular origin and the protease responsible for IFN-g<br />

proteolysis is most likely a metalloprotease (Molecular Weight close to<br />

90 kDa).<br />

Conclusions: Using zymogram analysis, gelatinase and caseinase<br />

activities in CHO batch cultures performed with or without serum were<br />

detected, and belong most likely to the metalloprotease and serine<br />

protease families. When cultures were carried out in RPMI with serum, a<br />

degradation of recombinant IFN-g was observed, while no IFN-g<br />

proteolysis was detected in culture performed with BDM medium.<br />

Furthermore, our results showed that despite the medium used (RPMI,<br />

BDM, with or without serum), addition of iron minimized IFN-g<br />

proteolysis, probably due to the inhibition of a 90 kDa metalloprotease<br />

activity. Thus, we demonstrated that the addition of iron citrate can be<br />

advantageously considered for industrial processes to prevent the<br />

proteolysis of a recombinant protein, in particular if one or several<br />

metalloproteases are present in the culture.<br />

Acknowledgements<br />

This work was financed by the Agence Nationale pour la Recherche<br />

Technique (ANRT) and Genclis SAS (Vandoeuvre-lès-Nancy, France).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P115) Western blot analysis of excreted IFN-g produced by CHO cells cultivated in various media; RPMI serum, BDM and RPMI serum<br />

supplemented with iron citrate.<br />

Table 1(abstract P115) Protease activities detected by zymography during CHO cell cultures performed in RPMI<br />

medium with serum and BDM medium<br />

Molecular Weight (kDa) RPMI medium with serum BDM medium<br />

Caseinase activities 220, 140, 90, 85 and 40 kDa 90 and 85 kDa<br />

Gelatinase activities 95, 90 and 65 kDa 90 kDa<br />

References<br />

1. Castro PML, Ison AP, Hayter PM, Bull AT: The macroheterogeneity of<br />

recombinant human interferon-g produced by Chinese-hamster ovary<br />

cells is affected by the protein and lipid content of the culture medium.<br />

Biotechnol Appl Biochem 1995, 21:87-100.<br />

2. Mols J, Peeters-Joris C, Wattiez R, Agathos SN, Schneider Y-J: Recombinant<br />

interferon-g secreted by Chinese hamster ovary-320 cells cultivated in<br />

Page 157 of 181<br />

suspension in protein-free media is protected against extracellular<br />

proteolysis by the expression of natural protease inhibitors and by the<br />

addition of plant protein hydrolysates to the culture medium. In Vitro<br />

Cell Dev- An 2005, 41:83-91.<br />

3. Schneider Y-J: Optimization of hybridoma cell growth and monoclonal<br />

antibody secretion in chemically defined serum- and protein-free<br />

medium. J Immunol Methods 1989, 116:67-77.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

P116<br />

Isolation of active peptides from plant hydrolysates that promote Vero<br />

cells growth in stirred cultures<br />

Samia Rourou, Rym Hssiki, Héla Kallel *<br />

Viral Vaccines Research & Development Unit, Institut Pasteur de Tunis, 13,<br />

place Pasteur. BP.74 1002 Tunis, Tunisia<br />

E-mail: Hela.Kallel@pasteur.rns.tn<br />

BMC Proceedings 2011, 5(Suppl 8):P116<br />

Background: Vero cells are adherent cell lines commonly used for the<br />

production of viral vaccines. We had developed an animal component<br />

free medium that allows an optimal growth of this cell line in stirred<br />

bioreactor [1,2]. We had also showed that Vero cells grown in this<br />

medium (called IPT-AFM) sustained rabies virus replication, and resulted<br />

in an overall yield comparable to the level obtained in serumsupplemented<br />

medium.<br />

IPT-AF medium contains plant hydrolysates, namely soy (Hypep 1510) and<br />

wheat gluten hydrolysates (Hypeps 4601 and 4605). These peptones were<br />

shown to promote cell attachment and growth. However, although these<br />

components are of non-animal origin, their use in vaccine production<br />

process has several drawbacks, mainly due variability between lots.<br />

The aim of this work is to identify active peptides from these hydrolysates<br />

that show a positive effect on cell adhesion, attachment and growth. For<br />

this purpose, the hydrolysates were fractionated using chromatography<br />

and precipitation techniques. The effect of the isolated fractions on Vero<br />

cells growth were tested in 24 and 6-well cell culture plates using<br />

experimental design approach. Fractions that sustain cell growth, were<br />

Figure 1(abstract P116) Vero cells on 2 g/l Cytodex 1 in spinner flask, in different media.<br />

Page 158 of 181<br />

further tested in stirred culture on Cytodex1 microcarriers, to confirm<br />

their positive effect on Vero cells growth.<br />

Materials and methods: Cell line: Vero cells adapted to IPT-AF medium<br />

as described in Rourou et al. [2] were used in this study.<br />

Media: M199 medium was purchased from Invitrogen, IPT-AFM was<br />

prepared as detailed in Rourou et al. [2]. Hypeps were provided by<br />

Sheffield Bio-Science.<br />

Culture systems: Static cultures: Cells were grown in 24-well plates<br />

(Falcon) and 6-well plate experiments « Nunclon Δ». The inoculation<br />

density was 2x10 5 cells/ml and the working volume was equal to 1 ml for<br />

the 24-well plate and 3 ml when the cells were grown in 6-well plate.<br />

Cells were grown at 37°C in 5% CO 2 incubator.<br />

Stirred cultures: Cells were grown in 6-well low binding plates (Costar)<br />

and in spinner flasks; cultures were inoculated at a cell density of 2x10 5<br />

cells/ml. The Working volume was equal to 3 ml for the 6-well plate and<br />

200 ml when cultures were performed in spinner flask. Cells were grown<br />

on 2 g/l Cytodex 1 at 37°C and 30 rpm.<br />

Fractionnation protocols: Hypeps 1510, 4601 and 4605 were fractionated<br />

by either chromatography methods or sequential precipitation with<br />

different ethanol concentrations as described in Shen et al. [3].<br />

Sephadex G-25 (GE Healthcare), BioGel P-2 fine (Biorad), Sephadex G-10<br />

(GE Heathcare), HiTrap Q HP (GE Healthcare) and HiTrap SP HP (GE<br />

Healthcare) matrixes were tested for the fractionation of the different<br />

solutions of Hypeps. Fractionation was monitored by measuring the<br />

absorbance of the collected fractions at 214 nm. Collected fractions were<br />

desalted, then frozen at -70°C and lyophilized. After lyophilization the<br />

fractions were resuspended in M199 so they would be compatible with<br />

cell culture.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Analytical methods: Cells grown in static cultures were first detached<br />

with the TrypLe Select (Invitrogen) then counted according to the Trypan<br />

blue method. Vero cells cultivated on Cytodex 1 microcarriers were<br />

counted using the Crystal Violet technique.<br />

Experimental design and statistical analysis: The software Modde 6.0<br />

(Umetrics, Sweden) was used in this study for the design of the<br />

experiments and the statistical analysis of the data.<br />

Results: Sephadex G-10, Sephadex G-25 and Biogel fine-2 were used to<br />

fractionate Hypeps 4605 and 4601. However, none of these matrixes was<br />

efficient.<br />

Anion and cation exchange chromatography were therefore used as an<br />

alternative method; two pH levels were tested: 5 and 8. Although these<br />

methods allowed the isolation of differentfractionsforeachpeptone,<br />

none of them had enhanced Vero cells when the cells were cultivated in<br />

24-well culture plates. In addition, most of these fractions showed a toxic<br />

effect on cell growth.<br />

Sequential precipitation with different ethanol concentration was also<br />

applied for the fractionation of the three peptones. The fractionation was<br />

conducted as described by Shen et al. [3]; 5 fractions were obtained for<br />

Hypep 4605 whereas for Hypep 4601 and Hypep 1510, 4 fractions were<br />

obtained for each. The effects of the isolated fractions on Vero cells growth<br />

were investigated in 24-well plates using a full factorial experimental<br />

design; 83 combinations were assessed in duplicate. Two combinations of<br />

fractions that show a cell growth comparable to that obtained in IPT-AF<br />

medium (positive control), were selected (combinations 1 and 2). The<br />

identified combinations were also tested in 6-well plates on 2 g/l Cytodex<br />

1 microcarriers. The highest cell density level reached under these<br />

conditions was similar to that achieved in IPT-AF medium.<br />

These combinations were further tested in spinner flask on Cytodex1<br />

microcarriers, to confirm their positive effect on Vero cells growth. Data<br />

shown in Figure 1, indicate that cell density level reached 2x10 6 cells/ml<br />

after 5 days of culture when Vero cells were grown in combination 1.<br />

Such level was slightly lower than that obtained in IPT-AF medium (2x10 6<br />

cells/ml versus 2.4x10 6 cells/ml). However, Vero cell growth in<br />

combination 2 was less efficient; the highest cell density obtained in this<br />

medium was equal to 1.7x10 6 cells/ml. Thus, combination 1 appears to<br />

be more suitable for Vero cells on Cytodex 1 microcarriers.<br />

Conclusions: Sephadex G-25, Sephadex G-10, Biogel-fine and ion<br />

exchange chromatography matrixes were not efficient for Hypeps<br />

fractionation. Sequential precipitation with ethanol appears to be the best<br />

method to isolate various fractions that promote Vero cells growth. Two<br />

Combinations (1 & 2) were identified as the best in terms of cell density<br />

and cell attachment. Spinner cultures demonstrated that these<br />

combinations are suitable for Vero cells growth on Cytodex 1. Further<br />

fractionation and characterization of these compounds are ongoing to<br />

identify the active components.<br />

Page 159 of 181<br />

References<br />

1. Rourou S, van der Ark A, Majoul S, Trabelsi K, van der Velden T, Kallel H: A<br />

novel animal component free medium for rabies virus production in<br />

Vero cells grown on Cytodex 1 microcarriers in a stirred bioreactor. Appl<br />

Microbiol Biotechnol 2009, 85:53-63.<br />

2. Rourou S, Van Der Ark A, Van Der Velden T, Kallel H: Development of an<br />

animal component free medium for Vero cells culture. Biotechnol Prog<br />

2009, 25:1752-1761.<br />

3. Shen CF, Kiyota T, Jardin B, Konishi Y, Kamen A: Characterization of<br />

yeastolate fractions that promote insect cell growth and recombinant<br />

protein production. Cytotechnol 2007, 54:25-34.<br />

P117<br />

Comparison of different membrane supports for monolayer culture of<br />

bovine oviduct epithelial cells<br />

Muhammad Z Tahir 1,2* , Fabien George 2 , Isabelle Donnay 2<br />

1 UMR-BDR 1198 INRA/ENVA, Ecole Nationale Vétérinaire d’Alfort, 94700<br />

Maisons-Alfort, France; 2 ISV-EMCA, Université Catholique de Louvain, 1348<br />

Louvain-la-Neuve, Belgium<br />

E-mail: mztahir@vet-alfort.fr<br />

BMC Proceedings 2011, 5(Suppl 8):P117<br />

Background: The oviduct epithelium consists of ciliated and secretory<br />

cells which play an important role in key reproductive processes such as<br />

sperm capacitation, fertilization and early embryonic development. Bovine<br />

oviduct epithelial cells have been widely used in co-culture experiments<br />

to condition culture media and improve early embryonic development<br />

[1]. However, these cells dedifferentiate during in vitro culture and<br />

manifest alterations like loss of cilia and secretory granules, reduction of<br />

cell height and flattening on the culture surface [2]. The trials for long<br />

term culture of oviduct cells, ensuring a better polarization and<br />

differentiation of the cells, include culture of oviduct cells on matrigel<br />

supports or collagen filter inserts. In this study, we have compared three<br />

different membrane supports for their potential to maintain<br />

ultrastructural features and monolayer integrity of bovine oviduct<br />

epithelial cells during in vitro culture.<br />

Materials and methods: Oviducts were excised from the genital tracts<br />

of cows slaughtered in abattoir. Following mechanical isolation, the<br />

oviduct epithelial cells were cultured on three different membrane<br />

supports i.e. polyester membrane (Thincert, Millipore Inc. USA),<br />

polytetrafluoroethylene membrane(Transwell, CorningInc.USA)and<br />

cellulose ester membrane (Millicell, Millipore Inc. USA). The culture of<br />

oviduct epithelial cells was done inTCM-199medium(Gibco,Grand<br />

Island, NY, USA) with 10% fetal bovine serum at 39°C in a humidified<br />

atmosphere of 5% CO 2 & 20% O 2. The development of primary cultures<br />

Figure 1(abstract P117) 1a) TEER (transepithelial electrical resistance) across oviduct cells monolayer (median ± extreme values) on 1. Thincert,<br />

2. Transwell, 3. Millicell inserts. 1b) Mannitol test (permeability to radioactive mannitol) through oviduct cells monolayer (median ± extreme values) on<br />

1. Thincert, 2. Transwell, 3. Millicell inserts.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

was assessed daily and the medium was renewed every 48 hours. After<br />

3 weeks of monolayer culture, the ultrastructural features of oviduct cells<br />

were examined by scanning electron microscopy while the integrity of<br />

monolayer was tested by transepithelial electrical resistance and<br />

permeability to radiolabeled 14 C-Mannitol. Non-parametric statistical<br />

analysis was done using Kurskal-Wallis Test.<br />

Results: The study of ultrastructural features by scanning electron<br />

microscopy revealed presence of an intact monolayer of polygonal<br />

epithelial cells on polyester (Thincert) and cellulose ester (Millicell)<br />

membrane supports while no such monolayer was seen in<br />

polytetrafluoroethylene (Transwell) membrane support. The test of<br />

transepithelial electrical resistance across monolayer of oviduct epithelial<br />

cells showed no significant difference (P>0.25) between the three<br />

membrane supports (Fig. 1a). The test for permeability of radiolabeled<br />

14 C-Mannitol across the monolayer of oviduct epithelial cells showed a<br />

tendency for lowest permeability (P=0.6) in polyester (Thincert)<br />

membrane support (Fig. 1b).<br />

Conclusion: The potential of polyester membrane supports (Thincert) to<br />

maintain ultrastructural features (intact monolayer of polygonal epithelial<br />

cells) and ensure monolayer integrity (higher transepithelial electrical<br />

resistance and lowest permeability to mannitol) during in vitro culture<br />

may serve as a model to study different cellular functions such as<br />

transport, absorption and secretory capacity of bovine oviduct epithelial<br />

cells as well as embryo-maternal interactions.<br />

References<br />

1. Rottmayer R, Ulbrich S, Kölle S, Prelle K, Neumüller C, Sinowatz F, Meyer H,<br />

Wolf E, Hiendleder S: A bovine oviduct epithelial cell suspension culture<br />

system suitable for studying embryo-maternal interactions:<br />

morphological and functional characterization. Reproduction 2006,<br />

132:637-648.<br />

2. Reischl J, Prelle K, Schöl H, Neumüller C, Einspanier R, Sinowatz F, Wolf E:<br />

Factors affecting proliferation and dedifferentiation of primary bovine<br />

oviduct epithelial cells in vitro. Cell Tiss Res 1999, 296:371-383.<br />

P118<br />

Efficacy of an inactivated and adjuvanted “ZULVAC® 8 OVIS” vaccine<br />

produced using single-use bioreactors<br />

Lídia Garcia * , Helena Paradell, Mercedes Mouriño, Berta Alberca, Alicia Urniza,<br />

Ana Vila, Margarita Tarrats, Joan Plana-Durán<br />

Pfizer Olot S.L.U., Ctra. Camprodon s/n, La Riba, 17813 Vall de Bianya<br />

(Girona), Spain<br />

E-mail: Lidia.garcia@pfizer.com<br />

BMC Proceedings 2011, 5(Suppl 8):P118<br />

Introduction: Bluetongue virus (BTV) first emerged in the European<br />

Union (EU) in 2006, peaking at 45,000 cases in 2008. The EU spent million<br />

of euros in 2008 and 2009 on eradicating and monitoring programs, cofinanced<br />

with member states. The number of cases in 2009 was 1118,<br />

with only 120 reported across the EU so far last year. Vaccination has<br />

proven itself as the most effective tool to control and prevent the disease<br />

and to facilitate the safe trade of live animals.<br />

Mammalian cells are commonly used as a substrate for production of<br />

most of the viral vaccines. BHK-21 cells are used for the production of<br />

bluetongue vaccines. Most companies use roller bottles or conventional<br />

bioreactors, but taking into account that the manufacturing cost is very<br />

important, the possibility of using Single-Use Bioreactor (SUB) technology<br />

as an alternative to roller bottles or conventional bioreactors was<br />

explored. Advantages like yields of production, time reduction<br />

Table 1(abstract P118) NA titers against BTV serotype 8 after vaccination<br />

(elimination of cleaning and sterilization steps needed for bioreactors, no<br />

validation process, etc) and quality of antigen production were studied.<br />

Materials and methods: Cell line: BHK-21 cell line was used due to the<br />

good yields. Cells were cultured at 37°C in Minimum Essential Medium<br />

Glasgow supplemented with serum.<br />

Cultivation system: The growth of the BHK-21 cells and virus production<br />

was conducted in roller bottles (RB), 10-L bioreactor (Biostat B-plus) and<br />

in 250-L SUB (Hyclone).<br />

Cells on bioreactors and SUB were cultivated using microcarriers<br />

(Cytodex-3) at a density of 3g/L. The crystal violet dye nucleus staining<br />

method was used to estimate cell density.<br />

Dissolved oxygen (DO) and pH were automatically adjusted by addition<br />

gases (CO 2, and air).<br />

Virus strain: BTV serotype 8 (BTV-8), strain BEL2006/02 was used in all<br />

experiments. The strain was supplied by “Veterinary and Agrochemical<br />

Research Centre” (VAR-CODA-CERVA), Ukkel, Belgium.<br />

Once the cells were 80-100% confluent, RB, 10-L bioreactor and 250-L<br />

SUB were infected under identical conditions and with a constant<br />

multiplicity of infection (MOI). Harvesting of virus was done when<br />

cytopathic effect (CPE) was about 90- 100%.<br />

Virus production was evaluated by TCID 50/ml.<br />

Vaccine: The antigen obtained on SUB was inactivated by Binary<br />

Ethylenimine (BEI) and adjuvanted with aluminum hydroxide and<br />

saponin. The efficacy of the vaccine was tested in lambs.<br />

Animals and experimental design: Forty, 1.5 month-old, lambs were<br />

included in the study.<br />

Thirty lambs were vaccinated and revaccinated 3 weeks later by<br />

subcutaneous route and ten lambs were left as unvaccinated controls.<br />

Forty-four days after revaccination all lambs were challenged with BTV-8.<br />

The antibody response in vaccinated lambs was evaluated from<br />

vaccination until the moment of challenge by a seroneutralization test<br />

(see table 1).<br />

Viremia (presence of BTV genome in blood samples) was evaluated by<br />

real time qRT-PCR [1] in blood samples obtained before challenge and<br />

during four weeks after challenge.<br />

Results and discussion: Some experiments to scale-up were done. An<br />

optimized and transferable process was developed in 10L glass vessels by<br />

monitoring the pH, temperature, and DO. The final process was<br />

transferred to 250-L stirred-tank SUB integrated with an Applikon<br />

controller.<br />

Cell growth and virus production in SUB was conducted at the optimal<br />

conditions determined previously on conventional bioreactors. Three<br />

critical parameters were taken into account: Cell concentration, growth<br />

rate at start virus inoculation and harvesting time.<br />

Cell growth: Several studies were conducted to compare the growth of<br />

cells in RB, 10-L bioreactor and 250-L SUB. During culture in bioreactors<br />

and SUB, DO was regulated by pulse of oxygen and pH was controlled.<br />

Figure 2, shows that cell concentration at 48 hours (when confluency is<br />

reached) was higher in bioreactors than in roller bottles. That<br />

corresponds to an increase of cell biomass by a factor of 1.5.<br />

Virus production: Figure 3, shows the results of the virus titers in 250-L<br />

SUB compared with those obtained in RB and 10-L bioreactor.<br />

The virus titer reached in a 10L bioreactor was comparable with the levels<br />

obtained in SUB. Preliminary results prove that by using SUB, the yields<br />

obtained in roller bottles can increase more than two times.<br />

Efficacy study: The potency of antigens produced using 250-L SUB<br />

technology was verified in target species.<br />

Serological study: Determination of neutralizing antibodies (NA) titer<br />

against BTV serotype 8 in the samples taken after vaccination and<br />

Mean NA Titers<br />

Group D


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P118) Scale-up from roller bottles to 250-L SUB.<br />

Figure 2(abstract P118) BHK-21 concentration at 48 hours post cultivation.<br />

Figure 3(abstract P118) Virus titers obtained at 72h p.i.<br />

Page 161 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

revaccination was done by means of seroneutralization test. Table 1 show<br />

the geometric mean NA titers.<br />

Evaluation of viremia after challenge: In any of the lambs of<br />

the vaccinated and challenged groups, viral gemone could be<br />

detected by RT-PCR during 27 days after challenge. Whereas in all<br />

unvaccinated and challenged lambs, the viral genome was detected<br />

from day 3-5 p.i.<br />

Conclusions: The results obtained using a 250-L SUB concerning the<br />

cell concentration and virus titer, were similar to those reached in a 10L<br />

bioreactor and higher than in roller bottles.<br />

Efficacy results on lambs confirm that the quality of antigen produced in<br />

SUB are similar of the antigen produced both in roller bottles and on<br />

conventional bioreactor.<br />

We can think on the possibility of using disposable systems in vaccines<br />

production in order to reduce the production costs.<br />

SUB can be an alternative to conventional production methods:<br />

Reduced facility complexity, reduction of the cost of building, and<br />

possibility of the rapid expansion of the capacity of the production.<br />

Reduction of the capital of equipment and equipment validation, etc.<br />

Avoid the cleaning process and reduction of the risk of crosscontamination.<br />

Reference<br />

1. Toussaint JF, Sailleau C, Breard E, Zientara S, De Clercq KJ: Bluetongue virus<br />

detection by two real-time RT-qPCRs targeting two different genomic<br />

segments. Virol Methods 2007, 140(1-2):115-123.<br />

P119<br />

Engineering CHO cell metabolism for growth in galactose<br />

Natalia E Jiménez 1,2 , Camila A Wilkens 1,2 , Ziomara P Gerdtzen 1,2*<br />

1 Centre for Biochemical Engineering and Biotechnology, Department of<br />

Chemical Engineering and Biotechnology, University of Chile, Santiago,<br />

8370448, Chile; 2 Millennium Institute for Cell Dynamics and Biotechnology: a<br />

Centre for Systems Biology, University of Chile, Santiago, 8370448, Chile<br />

E-mail: zgerdtze@ing.uchile.cl<br />

BMC Proceedings 2011, 5(Suppl 8):P119<br />

Background: Chinese hamster ovary (CHO) cells are one of the main<br />

hosts for industrial production of therapeutic proteins, owing to wellcharacterized<br />

technologies for gene transfection, amplification, and<br />

selection of high-producer clones. This has motivated the search for<br />

different strategies for the improvement of their specific productivity<br />

being one of the key points for this approaches the reduction of<br />

metabolic end-products like lactate and ammonia.<br />

Page 162 of 181<br />

The use of different carbon sources has been an alternative solution for<br />

this problem, as they are metabolized more slowly than glucose leading to<br />

lower production of metabolic end-products [1]. Particularly, it has been<br />

observed that cultures in presence of glucose and galactose undergo a<br />

metabolic shift in which they are capable of remetabolize lactate. However,<br />

the specific growth rate is diminished due to a slower metabolism<br />

associated to the incorporation of galactose [2]. In addition, cells are<br />

unable to survive with galactose as their unique carbon source [3].<br />

In this work we aim at identifying culture conditions that extend the<br />

culture’s viability for tPA producing CHO cells in media with combined<br />

glucose and galactose as carbon sources. Furthermore, we propose<br />

reducing the production of secondary metabolites by over-expressing<br />

galactokinase (GALK1), a bottleneck point in the galactose metabolism.<br />

Methodology: Two sets of experiments were carried out. In the first set,<br />

t-PA producing CHO TF 70R cells were grown in protein and serum free<br />

media without glutamine and supplemented with glucose, galactose and<br />

glutamate to define two different culture conditions. A high glucose<br />

control experiment was performed with 20 mM glucose (G20), and a<br />

combined carbon condition with a final concentration of 20 mM of<br />

hexose with 6 mM glucose and 14 mM galactose (GG6/14).<br />

Based on these results, transfection of genes involved in galactose<br />

metabolism are performed to enhance cell growth by improving<br />

galactose metabolism. Cells were transfected with the Galactokinase<br />

(GALK1) gene from Mus musculus, using Lipofectamine. Experiments were<br />

performed to analyze cell proliferation, carbon source consumption<br />

lactate production and metabolic flux distribution for the obtained<br />

pooled clones, as a preliminary tool to determine if the over-expression<br />

of this protein has a positive effect.<br />

In the second culture set, transfected cells were grown in protein free<br />

media with 2% Fetal Bovine Serum, supplemented with glucose and<br />

galactose to define two different culture conditions with a final<br />

concentration of 20 mM of hexose: 6 mM glucose and 14 mM galactose<br />

(GalKGG6/14); and 20 mM galactose (Gal20). Extracellular metabolites<br />

were measured.<br />

Results: In combined carbon source experiments, two phases can be<br />

distinguished. During the glucose consumption phase cells produce<br />

biomass, tPA, lactate and alanine. When glucose is depleted GG6/14<br />

culture enters a second metabolic stage where no significant growth is<br />

observed and galactose is consumed along with extracellular lactate and<br />

alanine. This is consistent with lactate and alanine being used as a<br />

supplementary pyruvate source to support energy metabolism associated<br />

with cellular maintenance.<br />

Metabolic flux analysis (MFA) was performed, considering the main<br />

reactions of the central glucose and galactose metabolism. Figure 1(a)<br />

Figure 1(abstract P119) Experimental results and metabolic flux distribution. (a) Metabolic Flux distribution between G20 (red) and GG6/14 (blue) at 50 h<br />

and 100 h in culture. Cell density, glucose and lactate concentration in GG6/14 (b) and Gal20 media (c). (▪) CHO tPA,(●) CHO tPA-pcDNA3.1(+), (♦) CHO<br />

tPA-GALK1, glucose (▪,♦), lactate (▲,●).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

shows results for two time-points for each culture, an early (50 h) and a<br />

late time-point (100 h). For GG6/14, the columns represent glucose and<br />

galactose consumption stages, respectively. In the pyruvate node carbon<br />

molecules are channeled either towards the TCA cycle, or in the direction<br />

of secondary metabolite synthesis such as lactate and alanine. Fluxes<br />

from the central carbon metabolism are reduced several times in the late<br />

time point for both cultures, except for the Pyr-AcCoA reaction, which<br />

plays a central role in the regulation of mammalian metabolism by<br />

connecting glycolysis with the TCA cycle. In late culture stages where<br />

hexose uptake is low, cell metabolism is directed towards maintaining<br />

TCA cycle fluxes in order to obtain energy. To achieve this in GG6/14,<br />

galactose and lactate are used as an additional carbon source.<br />

SincegalactoseconsumptionappearstobelimitingcellgrowthinGG6/<br />

14, there is an open possibility to improve cell growth during galactose<br />

consumption while maintaining low lactate production by overexpressing<br />

genes involved in galactose metabolism. Transport of<br />

galactose has been proposed to explain the low uptake of galactose and<br />

the GalK reaction is identified as the limiting step of the galactose<br />

metabolism. To achieve this cells are transfected with stable expression<br />

vectors to insert the galactokinase GalK1 gene and the galactose<br />

transporter Slc2a8.<br />

Obtained clones exhibit a higher growth rate in galactose than control<br />

cells due to their ability of metabolize galactose at a increased rate.<br />

Transfected cells in GalKGG6/14 consume all available glucose before<br />

starting to consume galactose. They also exhibit lower lactate<br />

accumulation, indicating that the introduction of these gene does not<br />

increase galactose uptake in a manner that would generate secondary<br />

metabolites. Control cells are unable to grow on media with galactose as<br />

the only carbon source. Transfected cells were able to maintain high<br />

viability during 100 hours in this media.<br />

The over-expression of the Slc2a8 (GLUT8) Galactose transporter could<br />

enhance the positive effects of the introduction of the galactokinase<br />

gene by increasing the availability of substrate at an intracelullar level.<br />

Before transfection, mutation of a dileucine motif to alanine is required to<br />

allow the expression of this transporter in the plasmatic membrane. For<br />

that purpose an strategy using PCR with primers that include this<br />

substitutions is currently undergoing.<br />

Conclusions: When CHO cells are cultured with a combination of glucose<br />

and galactose it is possible to achieve extended viability along with a<br />

metabolic shift towards lactate consumption, which is triggered when the<br />

second hexose is consumed.<br />

Metabolism can be modified through cell engineering to increase cell<br />

growth while maintaining low lactate production rates, enabling cells to<br />

utilize alternative carbon sources. Specifically this can be achieved with<br />

galactose.<br />

Acknowledgements: This work was supported by FONDECYT Initiation<br />

Grant 11090268.<br />

References<br />

1. Wlaschin K, Hu WS: Engineering cell metabolism for high density cell<br />

culture via manipulation of sugar transport. J of Biotechnol 2007,<br />

13:1023-1027.<br />

2. Altamirano C, Illanes A, Becerra S, Cairó J, Gòdia F: Considerations on the<br />

lactate consumption by CHO cells in the presence of galactose. Jof<br />

Biotechnol 2006, 125:547-556.<br />

3. Neerman J, Wagner R: Comparative analysis of glucose and glutamine<br />

metabolism in transformed mammalian cell lines, insect and primary<br />

liver cells. J Cell Physiol 1996, 166:152-169.<br />

P120<br />

Engineering CHO cells for improved central carbon and energy<br />

metabolism<br />

Camila A Wilkens 1,2 , Ziomara P Gerdtzen 1,2*<br />

1 Centre for Biochemical Engineering and Biotechnology, Department of<br />

Chemical Engineering and Biotechnology, University of Chile, Santiago,<br />

8370448, Chile; 2 Millennium Institute for Cell Dynamics and Biotechnology: a<br />

Centre for Systems Biology, University of Chile, Santiago, 8370448, Chile<br />

E-mail: zgerdtze@ing.uchile.cl<br />

BMC Proceedings 2011, 5(Suppl 8):P120<br />

Background: Investigations have shown animal cell cultures’<br />

performance, in terms of cell proliferation and production of recombinant<br />

Page 163 of 181<br />

protein, are negatively affected by both lactate’s concentration and its<br />

specific production rate. In a previous work, we determined that lactate<br />

production was caused by pyruvate accumulation due to its high<br />

synthesis rate in the glycolitic pathway and limited consumption in the<br />

TCA cycle, which leads to lactate production [1]. In this work, we use the<br />

ΔL/ΔHexose ratio in order to characterize the cells metabolic state. This<br />

ratio describes the lactate production rate vs. hexose consumption. Low<br />

ΔL/ΔHexose ratios indicate efficient metabolic states where carbons<br />

consumed are mainly used to support cell growth, protein synthesis or<br />

energy metabolism.<br />

Cell engineering has been previously used to improve cultures’<br />

performance by changing the expression of genes involved in<br />

metabolism and apoptosis, focusing on the modification of only one<br />

gene at the time. These works showed that after overexpression of genes<br />

such as fructose transporter (Slc2a5) and yeast’s pyruvate carboxylase<br />

(PYC) cells are able to achieve higher cell densities and lower lactate<br />

production than wild-type cells under the same culture conditions [2-4].<br />

In this work we aim at introducing multiple changes in the cells’ genome<br />

in order to obtain an engineered cell line with reduced lactate<br />

production and enhanced energy metabolism, which is capable of<br />

achieving higher cell densities and with a longer lifespan. We propose to<br />

control both, carbon uptake and itsusebytheTCAcycle.Cellswere<br />

transfected with the fructose transporter gene (Slc2a5) and pyruvate<br />

carboxylase gene (PYC). Metabolic flux redistribution was studied through<br />

metabolic flux analysis, comparing engineered cells and wild-type under<br />

normal culture conditions.<br />

Materials and methods: CHO cells were transfected with the pcDNA3.1<br />

(+) zeo-Slc2a5 and/or PCMVSHE-PYC2 + Hygromycine resistance vectors<br />

using lipofectamine. After selection, five experiments were designed to<br />

study cell proliferation, carbon source consumption, lactate production<br />

and metabolic fluxes. CHO cells overexpressing PYC (CHO-PYC) were<br />

cultured with glucose 17.5 mM and cells transfected with Slc2a5 (CHO-<br />

Slc2a5) and both PYC and Slc2a5 (CHO-PYC-Slc2a5) were grown in media<br />

containing fructose 17.5 mM. Two control cultures were performed with<br />

wild-type CHO cells in 17.5 mM glucose (GC) orfructose(FC). Results are<br />

shown in Figure 1.<br />

Results: Cultures’ performance: As seen in Figure 1.(a) and Table 1<br />

respectively, GC, CHO-PYC and CHO-PYC-Slc2a5 were able to reach higher<br />

cell densities and maximum growth rates (µ max) than FC and CHO-Slc2a5.<br />

Cultures with glucose have almost no lag phase while experiments with<br />

media supplemented with fructose have long lag phases, probably due<br />

tothesloweruptakeoffructose,which would delay the exponential<br />

growth phase. In addition, engineered cells exhibit an extended lifespan<br />

in comparison to wild-type cells.<br />

ΔL/ΔHexose values reached by the cultures are given in Table 1. CHO cells<br />

grown in high glucose show an inefficient metabolic state where most<br />

carbons consumed go towards lactate production. Engineered cells<br />

grown in glucose have lower lactate production per carbon consumed<br />

than wild-type cells (Figure 1.(b)).<br />

Engineered cells show a better use of glucose, producing less lactate per<br />

glucose consumed, as reflected in their lower ΔL/ΔHexose. In addition,<br />

CHO-PYCcellsareabletoproduceless lactate and achieve a longer<br />

lifespan than wild-type cells. CHO-Slc2a5 cells have higher fructose<br />

uptake rates than FC and are able to achieve longer lifespans and higher<br />

cell densities.<br />

CHO-PYC-Slc2a5 cells have the highest µmax among all experiments yet<br />

they produce more lactate than FC. Most of the lactate is produced in<br />

the lag phase. The fact that cells are capable of growing in fructose as<br />

well as in glucose and have a better ΔL/ΔHexose than GC, indicates that<br />

there is room for further improvement of this system.<br />

Metabolic flux analysis: Figure 1.(c) shows the flux distribution of the<br />

different cultures for central carbon metabolism during mid exponential<br />

growth phase. CHO cells grown in fructose have lower amounts of<br />

carbon directed towards energy metabolism. Both CHO-Slc2a5 and CHO-<br />

PYC-Slc2a5 have higher fluxes in glycolysis and TCA cycle than FC,<br />

consistent with higher cell density. CHO-PYC cells consume lower<br />

amounts of glucose than GC, and most of it is directed towards the TCA<br />

cycle. CHO-Slc2a5 cells consume higher amounts of fructose than FC and<br />

most of it is directed towards the TCA cycle. CHO-PYC-Slc2a5 cells show a<br />

more active metabolism than FC, consuming more fructose, with higher<br />

TCA cycle fluxes and lactate production, while reaching higher cell<br />

densities than the control.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P120) Experimental and MFA results. Pink circles: GC, orangesquares:FC, purple upwards triangle: CHO-PYC , green downwards<br />

triangle: CHO-Slc2a5 , blue rhombus: CHO-PYC-Slc2a5. (a) Cell density, (b) Lactate concentration (c) Comparison of metabolic flux distribution in carbon<br />

mmol/10 9. cells/hr for the different experiments during mid exponential growth. Scale is the same in all graphs.<br />

Conclusions: It is possible to modify cells for a more efficient metabolism<br />

in media supplemented with glucose and fructose using cell engineering.<br />

Engineered cells show enhanced viability and more efficient metabolic<br />

states under high glucose or fructose concentrations than the controls.<br />

Acknowledgements<br />

We would like to thank Dr. Roland Wagner for the PCMVSHE-PYC2 vector,<br />

Dr. Mariella Bollati for the pcDNA3.1(+)zeo vector and the Genetically<br />

EngineeredMouseFacilityatM.D.AndersonCancerCenterforthe<br />

Hygromycine resistance cassette. This work was supported by FONDECYT<br />

Initiation Grant 11090268.<br />

References<br />

1. Wilkens CA, Altamirano C, Gerdtzen ZP: Comparative metabolic analysis of<br />

lactate for CHO cells in glucose and galactose. Biotechnology and<br />

Bioprocess Engineering Journal 2011 in press.<br />

2. Elias CB, Carpentier E, Durocher Y, Bisson L, Wagner R, Kamen A: Improving<br />

glucose and glutamine metabolism of human HEK 293 and Trichoplusia<br />

Table 1(abstract P120) Parameters for cell growth and<br />

ΔL/ΔHexose<br />

Experiment µ max [10 -2 hrs -1 ] ΔL/ΔHexose<br />

GC 1.63 1.7<br />

FC 0.86 0.81<br />

CHO-PYC 2.1 0.81<br />

CHO-Slc2a5 0.65 0.88<br />

CHO-PYC-Slc2a5 3.68 1.1<br />

Page 164 of 181<br />

ni insect cells engineered to express a cytosolic pyruvate carboxylase<br />

enzyme. Biotechnol Prog 2003, 19(1):90-97.<br />

3. Irani N, Wirth M, van Den Heuvel J, Wagner R: Improvement of the<br />

primary metabolism of cell cultures by introducing a new cytoplasmic<br />

pyruvate carboxylase reaction. Biotechnol Bioeng 1999, 66(4):238-246.<br />

4. Wlaschin KF, Hu WS: Engineering cell metabolism for high-density cell<br />

culture via manipulation of sugar transport. J Biotechnol 2007,<br />

131(2):168-176.<br />

P121<br />

Mitogenic effect of sericin on mammalian cells<br />

Wataru Sato 1* , Ken Fukumoto 1 , Kana Yanagihara 1 , Masahiro Sasaki 2 ,<br />

Yoshihiro Kunitomi 2 , Satoshi Terada 1<br />

1 Department of Applied Chemistry and Biotechnology, University of Fukui,<br />

Fukui, 910-8507, Japan; 2 Development Department, SEIREN Co., Ltd., Fukui,<br />

913-0038, Japan<br />

BMC Proceedings 2011, 5(Suppl 8):P121<br />

Fetal bovine serum (FBS) or other mammal-derived factors are extensively<br />

supplemented as growth factor into culture medium. Since mammalderived<br />

factors arouses the concern about the risk of zoonosis, serumand<br />

mammal-free culture is strongly required. We focused on sericin<br />

hydrolysates, originating from silkworm, and reported that sericin is<br />

effective as growth factor to various cells. But it is not elucidated how<br />

sericin induces the proliferation and inhibits apoptosis of the cells. In this<br />

study, inhibitor assay was done in order to identify signaling factors


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P121) The pathway deduced from our previous study. cDNA microarray analysis detected up-regulation of myc, tbp, fos, and Bcl-xL,<br />

and down-regulation of atf3 after sericin treatment. Inhibition assay confirmed the involvement of Ras and MEK1/2 in the mitogenic effect of sericin.<br />

Figure 2(abstract P121) Different pathways depending on cell line.<br />

Page 165 of 181


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

involved in sericin effect. In hybridoma cells, the involvement of Src was<br />

suggested, while ERK1/2, PP2A and p38 were not involved. In HepG2 cells,<br />

the involvement of Src and ERK1/2 was suggested. From these results, it<br />

was implied that the mitogenic effect of sericin might be transduced<br />

through different pathways depending on cell line.<br />

Background: In in vitro cell culture, mammal-derived factors including<br />

FBS are usually used as growth factors and supplemented into the media.<br />

However, supplementing mammal-derived factors causes the concern<br />

about the risk of zoonosis such as abnormal prions and various viruses.<br />

Therefore, mammal-free culture is strongly required in the industry of<br />

antibody therapeutics production and in regenerative medicine including<br />

cell therapy. As an alternative to mammal-derived factors, we focused on<br />

hydrolysate of sericin, which is glue protein included in silk fiber. Our<br />

previous studies revealed that sericin has mitogenic and anti-apoptotic<br />

effect on various cell lines [1-3]. Additionally, sericin hydrolysates treated<br />

with autoclave sterilization maintained its original mitogeneic activity and<br />

so we successfully developed a mammal-free medium, Sericin-GIT (Wako<br />

Pure Chemical Industries, Ltd., Japan). Although sericin is effective, it is<br />

not revealed how sericin up-regulates the proliferation and downregulates<br />

apoptosis.<br />

In the previous study, the following results were obtained. One, cDNA<br />

microarray analysis detected up-regulation of myc, tbp, fos and Bcl-xL, and<br />

down-regulation of atf3 after sericin treatment. Two, Inhibition assay<br />

confirmed the involvement of Ras and MEK1/2 in the mitogenic effect of<br />

sericin. From these results, we suppose a following pathway (Figure 1). In<br />

this study, inhibitor assays against the factors shown in the figure were<br />

done in order to identify signaling factors involved in sericin effect.<br />

Materials and methods: Cells. Mouse hybridoma 2E3-O cells and human<br />

hepatoblastoma HepG2 cells were cultured in ASF104 (Ajinomoto Japan)<br />

serum-free medium and DME medium (Nissui, Japan) without FBS,<br />

respectively.<br />

Inhibitors. PP2 (BioMoL, USA), ERK Activation Inhibitor 1, Cell-Permeable<br />

(Calbiochem, USA), Okadaic acid (Calbiochem), SB239063 (SIGMA, USA)<br />

were used at inhibition assay.<br />

Inhibition Assays. Mouse hybridoma 2E3-O cells and human<br />

hepatoblastoma HepG2 cells were cultivated in addition of each of<br />

inhibitors. After two days, the number of cells and the viability were<br />

determined by trypan blue-exclusion test with hemocytometer.<br />

Results: Inhibition assays were done in order to identify signaling factors<br />

involved in sericin effect.<br />

Since Ras and MEK1/2 are involved in Map kinase pathway, PP2, a specific<br />

inhibitor against Src, was used. PP2 successfully neutralized the mitogenic<br />

effect of sericin, indicating that Src would be involved.<br />

Involvement of ERK1/2 was tested by using ERK Activation Inhibitor 1. The<br />

inhibitor neutralized the mitogenic effect in HepG2 cells, but did not in<br />

hybridoma cells, suggesting that ERK1/2 would be involved in HepG2<br />

cells but not in hybridoma cells. In order to confirm that ERK1/2 is not<br />

involved in hybridoma cells, inhibition assay against PP2A was done; the<br />

mitogenic effect could be enhanced by inhibition of PP2A if MAPK<br />

pathway would be involved in the mitogenic effect of sericin. Inhibition<br />

against PP2A failed to improve the proliferation of the hybridoma cells<br />

treated with sericin, implying that PP2A would not be involved.<br />

Further cDNA microarray analysis implied that several other genes might<br />

be affected by sericin treatment. Among the genes, myc and stat1 are the<br />

lower signaling factors of p38 and so SB239063, a specific inhibitor<br />

against p38, was tested. It failed to neutralize, indicating that p38 would<br />

not be involved in the mitogenic effect of sericin.<br />

Conclusions: In hybridoma cells, Src was involved in the mitogenic effect<br />

of sericin, while ERK1/2, PP2A and p38 were not, suggesting that Src could<br />

activate other factors to transduce signal of sericin. In HepG2 cells, the<br />

involvement of Src and ERK1/2 was confirmed, suggesting that MAPK<br />

pathway could be involved. From these results, the mitogenic effect of<br />

sericin is transduced through different pathways depending on cell lines<br />

as shown in figure 2. Further study should be done to reveal the<br />

involvement of these factors.<br />

References<br />

1. Morikawa M, Kimura T, Murakami M, Katayama K, Terada S, Yamaguchi A:<br />

Rat islet culture in serum-free medium containing silk protein sericin.<br />

J Hepatobiliary Pancreat Surg 2009, 16:223-228.<br />

2. Yanagihara K, Terada S, Miki M, Sasaki M, Yamada H: Effect of the silk<br />

protein sericin on the production of adenovirus-based gene-therapy<br />

vectors. Biotechnol Appl Biochem 2006, 45:59-64.<br />

Page 166 of 181<br />

3. Terada S, Sasaki M, Yanagihara K, Yamada H: Preparation of silk protein<br />

sericin as mitogenic factor for better mammalian cell culture. J Biosci<br />

Bioeng 2005, 100:667-671.<br />

P122<br />

Challenges in scaling up a perfusion process<br />

Vana Raja, Saravanan Desan, Ankur Bhatnagar * , Anuj Goel, Harish Iyer<br />

Cell Culture Lab, Biocon Limited, Bangalore, India<br />

E-mail: ankur.bhatnagar@biocon.com<br />

BMC Proceedings 2011, 5(Suppl 8):P122<br />

Introduction: Perfusion process involves retention of the cells inside the<br />

bioreactor while simultaneously removing spent medium and adding<br />

fresh medium continuously. The flow rate of addition of fresh and<br />

removal of spent medium are generally kept the same (perfusion rate) to<br />

maintain the culture volume inside the bioreactor. Cell retention is<br />

possible with many devices but the reliability and consistency in<br />

performance of these devices remains a major challenge for design,<br />

operation and scale-up of perfusion processes. For a filtration based<br />

retention device, successful scale up depends on cell retention efficiency,<br />

prevention of filter fouling and the similarity of perfusion equipment<br />

between small and large scale. We present a case study where a<br />

perfusion process from a 2L (Liter) bioreactor is scaled up to<br />

manufacturing scale of 1KL (Kilo Litre).<br />

Materials and methods: A NS0 host cell line cultured in protein free<br />

medium was used. At lab scale 2L stirred tank bioreactors with internal<br />

spin filters as the retention device were used. The 1KL production<br />

bioreactor used closed external rotating filters for cell retention. Cell<br />

count and viability were determined using Hemocytometer and Trypan<br />

Blue dye exclusion. Glucose and lactate were measured using YSI 2700<br />

analyzer and product concentration by Affinity chromatography.<br />

Results and discussions: Development of perfusion process and<br />

scale up: A perfusion process was developed which consisted of a batch<br />

phase for cell growth followed by perfusion phase once the cell density<br />

reaches the desired level. This process was scaled up by scaling up the<br />

scale dependent parameters linearly same while maintaining the scale<br />

independent factors within the acceptable range. The filter parameters<br />

such as mesh type and filter area per unit bioreactor volume were<br />

however not comparable between the scales.<br />

PB-1 (Production Batch no. 1): The growth rate during the batch phase<br />

of the run was comparable to the lab scale. However, during the<br />

perfusion phase, the maximum cell densitywasobservedtobeonly<br />

about 25% of the lab scale. Due to the lower cell counts, the perfusion<br />

rates were also proportionally reduced. Together this resulted in<br />

obtaining only ~25% of the expected product yield.<br />

As part of investigation, the following factors were analyzed:<br />

a) Inoculum and “medium lot” used in PB-1 – A control batch was run in<br />

the lab with the same inoculum and medium as used in the PB-1 run.<br />

This batch showed normal lab batch profiles ruling out these factors as<br />

possible cause for the underperformance of the production batch.<br />

b) Pumps used for perfusion – The lab scale used peristaltic pumps while<br />

in manufacturing pulsating pumps were used. These pulsating pumps<br />

could influence cell retention and hence were replaced with peristaltic<br />

pumps for future production batches.<br />

PB-2 (Production Batch no. 2): Changing the pump type resulted in<br />

better cell retention. However, soon the retention started dropping,<br />

indicating cell loss from the bioreactor. Visual inspection of the filter<br />

mesh (mesh Type A) showed significant mesh deformation. This could<br />

have resulted due to the fragile nature of the mesh which could not<br />

withstand the negative pressure generated inside the closed filter<br />

housing due to suction forces and filter fouling. The mesh Type A was<br />

also found to be fouling very fast due to it mesh weave design. Two<br />

other mesh types (B & C) with different mesh weave patterns and<br />

mechanical strengths were evaluated. Type B showed poor cell retention<br />

due to bigger pore size. Type C showed better cell retention and also did<br />

not deform easily during filter fouling. Thus the filter mesh was changed<br />

from Type A to Type C in the production system.<br />

PB-3 (Production Batch no. 3): The filter with mesh C showed cell<br />

retentionbetterthaneventhelabscale.Thisresultedinachievingcell<br />

densities higher than the range of the lab batches (Figure 1). The<br />

perfusion rates were increased by about 20% from the lab scale to


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P122) Cell count profiles from the lab batches and the production batches with different mesh types.<br />

address the nutritional requirements of the higher cell numbers. The<br />

product yield was also proportionally higher by ~20%.<br />

Note: N. VCC – normalized viable cell concentration<br />

Product quality analysis: Analysis showed that the product obtained<br />

from batch PB-3 was significantly different in quality (charge distribution)<br />

compared to product from PB-1,2 and lab batches. The following were<br />

evaluated as possible reasons for the observed differences:<br />

a) High perfusion rates compared to lab and PB-1,2 batches resulting in<br />

shorter product residence time inside the bioreactor.<br />

b) Different lactate and pCO2 levels maintained (due to higher perfusion<br />

rates) which also resulted in different pH profiles.<br />

PB-4 (Production Batch no. 4): Batch PB-4 was run with perfusion flow<br />

rates comparable to the lab batches. The additional nutritional<br />

requirement for the higher cell concentration was addressed by making<br />

the fresh medium more concentrated. pH was also controlled using the<br />

CO 2 and base combination. The cell concentration obtained was similar<br />

to PB-3. Lowering the perfusion flow rates also helped in delaying the<br />

clogging of the filter.<br />

Product analysis showed that the changes done in the batch helped in<br />

bringing the product quality closer to (within acceptable range) the lab<br />

batches. The results are shown in the Table 1. Two differently charged<br />

species Type-1 and Type-2 are used for comparison.<br />

Table 1(abstract P122) Product quality comparison of PB-3<br />

and PB-4 batches<br />

Perfusion lots PB-3 PB-4<br />

Type-1* Type-2* Type-1* Type-2*<br />

1 112 74 59 139<br />

2 47 184 66 126<br />

3 43 195 73 116<br />

4 41 198 73 120<br />

5 39 208 82 109<br />

6 39 209 82 118<br />

* % of Lab batch profiles.<br />

Page 167 of 181<br />

Summary: Development and scale-up of a perfusion process has challenges<br />

due to the complex nature of the process and unavailability of direct scale-up<br />

of the perfusion equipment. The initial scale-up to production scale resulted<br />

in poor cell growth profile. Upon investigation, the reason for low cell<br />

concentration was attributed to poor cell retention by the perfusion device.<br />

Changes were introduced in the type of mesh used for filter construction and<br />

perfusion pumps to improve retention. These modifications helped in better<br />

cell culture profiles and yields. However, the product quality was impacted<br />

because of these changes. Further changes in the perfusion flow rates were<br />

done to address the product quality differences.<br />

P123<br />

High cell density growth of High Five suspension cells in DO-controlled<br />

wave-mixed bioreactors<br />

Teddy Beltrametti 1 , Nicole C Bögli 1* , Gerhard Greller 2 , Regine Eibl 1 , Dieter Eibl 1<br />

1 Institute of Biotechnology, Zurich University of Applied Sciences and Facility<br />

Management (ZHAW), Wädenswil CH-8820, Switzerland; 2 Sartorius Stedim<br />

Biotech, Göttingen, D-37075, Germany<br />

BMC Proceedings 2011, 5(Suppl 8):P123<br />

Insect cells such as High Five cells used in the manufacture of<br />

biopharmaceuticals are best grown in wave-mixed bioreactors (1). This is<br />

due to the continual blending of foam with the culture medium which<br />

results from the wave-induced mixing and permanent renewal of the<br />

medium surface. Even the addition of an antifoam agent is not required.<br />

Process conditions which ensure maximum High Five cell densities and<br />

whichhavebeenreportedtobeabout8x10 6 cells x mL -1 (2) were<br />

determined in Biostat CultiBag RM50 optical experiments for batch mode<br />

and 1 L culture volume. Seed inoculum for these experiments was generated<br />

in single-use shake flasks (Corning) incubated in an Infors`Multitron shaker<br />

(27°C, 100 rpm, 25 mm shaking diameter). Biostat CultiBag RM50 optical was<br />

controlled in four different modes: non-pH- and non-DO-controlled, DOcontrolled,<br />

pH-controlled, DO- as well as pH-controlled. The DO level was<br />

guaranteed by increasing the rocking rate up to 28 rpm and, if required by<br />

addition of pure oxygen. In process control was supplemented with off-line<br />

analyses of cell density, viability, metabolites (glucose, glutamine, glutamate,<br />

lactate, ammonium) and pH.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

While the influence of the type of bioreactor`s control on the maximum<br />

growth rate (0.041-0.044 h -1 ) and doubling time (15.6-17.7 h) was<br />

negligible, maximum cell densities were achieved with DO regulation (set<br />

point 50%). Maximum cell densities ranged between 8.2 and 9.4 x 10 6<br />

cells x mL -1 and represent the highest values described for High Five cells<br />

so far in the literature. They are 35% higher compared to those seen in<br />

pH-controlled and non-controlled experiments. Controlling both DO and<br />

pH level did not lead to any further improvement of cell growth i.e. the<br />

range of growth parameter values was the same as that observed in the<br />

previous experiments. For High Five cell-based biopharmaceuticals this<br />

knowledge enables optimized seed inoculum/seed train production in<br />

wave-mixed bag bioreactors.<br />

References<br />

1. Werner S, et al: Innovative, Non-stirred Bioreactors in Scales from Millilitres<br />

up to 1000 Liters for Suspension Cultures of Cells using Disposable Bags<br />

and Containers – A Swiss Contribution. CHIMIA 2010, 64:819-823.<br />

2. Rhiel M, Mitchell-Logean CM, Murhammer DW: Comparison of Trichoplusia<br />

ni BTI-Tn-5B1-4 (High FiveTM) and Spodoptera frugiperda Sf-9 Insect<br />

Cell Line Metabolism in Suspension Cultures. Biotechnology and<br />

Bioengineering 1997, 55:909-920.<br />

P124<br />

Bag-based rapid and safe seed-train expansion method for Trichoplusia<br />

ni suspension cells<br />

Nicole C Bögli 1* , Christoph Ries 1 , Irina Bauer 1 , Thorsten Adams 2 ,<br />

Gerhard Greller 2 , Regine Eibl 1 , Dieter Eibl 1<br />

1 Institute of Biotechnology, Zurich University of Applied Sciences and Facility<br />

Management (ZHAW), Wädenswil CH-8820, Switzerland; 2 Sartorius Stedim<br />

Biotech, Göttingen, D-37075, Germany<br />

BMC Proceedings 2011, 5(Suppl 8):P124<br />

Trichoplusia ni suspension cells (High Five) used in conjunction with<br />

the baculovirus vector expression system (BEVS) are regarded as<br />

potential product system of new, recombinant virus-like particle (VLP)<br />

vaccines. In order to push vaccine development and production,<br />

biomanufacturers use single-use technology when- and wherever<br />

possible. This applies to upstream processing and in particular seedtrain<br />

expansion ranging from cryopreserved vials via t-flasks, spinners<br />

(respectively shake flasks) to stirred stainless steel bioreactors. The<br />

stainless steel bioreactors deliver inoculum for seed bioreactors and<br />

have been increasingly replaced by wave-mixed single-use bag<br />

bioreactors during the last 5 years [1].<br />

Page 168 of 181<br />

The approach presented for seed-train cell expansion of High Five<br />

suspension cells is based on the Biostat CultiBag RM50 optical (Sartorius<br />

Stedim Biotech). It was used for the production of cells for long-term<br />

storage and for the expansion of cells for subsequent production<br />

experiments. For long-term storage the cells were frozen at high cell<br />

concentrations (20 - 40 x 10 6 cells x mL -1 ) in 60 mL Cryobags and stored<br />

in nitrogen at -196 °C in vapour phase.<br />

Initial experiments were aimed at the growth characterization of High Five<br />

suspension cells from a vial working cell bank (WCB). The High Five cells<br />

were grown in batch mode and in 250 mL single-use shake flasks (Corning<br />

and Sartorius Stedim Biotech) on a Certomat® CT Plus shaker (Sartorius<br />

Stedim Biotech) during six days (triplicates, 27 °C, 100 rpm, 25 mm shaking<br />

diameter). Afterwards a procedure was developed in which thawed cells<br />

from a Cryobag were directly transferred into and expanded in a Biostat<br />

CultiBag RM. Under optimal process conditions (500 mL starting volume, a<br />

starting cell density of 1 x 10 6 cells x mL -1 , 27 °C, rocking angle of 6 °, 20 -<br />

30 rpm, 0.2 vvm, DO set point 50%) growth rate (0.039 - 0.042 h -1 ),<br />

doubling time (18 - 20 h) and maximal cell density (7.8 - 8.9 x 10 6 cells x<br />

mL -1 ) showed good correlation with results arising from CultiBags which<br />

were inoculated with cells from shake flasks. This bag-based seed-train<br />

expansion allows time saving of about one week and reduces crosscontamination,<br />

both advantages being due to omitted intermediate<br />

cultivation steps in shake flasks.<br />

Reference<br />

1. Eibl R, Löffelholz C, Eibl D: Single-Use Bioreactors-An Overview. Single-Use<br />

Technology in Biopharmaceutical Manufacture Eibl R, Eibl D. Wiley , 1 2010,<br />

1:33-51.<br />

P125<br />

On-line monitoring of the live cell concentration in bioreactors based<br />

on a rocking platform<br />

John Carvell * , Matt Lee<br />

Aber Instruments Ltd., Aberystwyth, SY23 3AH, UK<br />

E-mail: johnc@aberinstruments.com<br />

BMC Proceedings 2011, 5(Suppl 8):P125<br />

Background: Aber Instruments first introduced the concept of using<br />

disposable live-biomass probes in 2009 [1]. The probe has been carefully<br />

designed to be welded into most single use bioreactors and is suitable<br />

for bags with agitators eg the Hyclone SUB and the Sartorius Stedim<br />

Biostat Cultibag STR, or those using the rocker type platform eg Sartorius<br />

Stedim Biostat Cultibag RM and GE Wave bioreactors . The disposable<br />

Figure 1(abstract P125) Capacitance and conductivity measurements in a 50-L Sartorius (25 L working volume) rocking-motion CultiBag system<br />

(R. Tanner, GlaxoSmithKline, UK)


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

biomass probe has four electrodes with the same dimensions as the<br />

existing reusable production biomass probes with flush platinum<br />

electrodes that are used in cGMP manufacture worldwide. The electrode<br />

support material is HDPE that meets FDA and USP Class VI requirements<br />

and the probe can withstand gamma sterilisation and be stored for<br />

prolonged periods before use. The disposable probe is easily connected<br />

to a mini-lightweight Futura pre-amplifier so that the weight load or<br />

torque on the bag is minimal and the bulk of the electronics is then<br />

located well away from the bag. The disposable probe has already been<br />

welded into different bags including Sartorius Stedim Biotech CultiBag<br />

RM bioreactors.<br />

Materials and methods: Experiments were performed with a Futura MRF<br />

Mini-Remote Futura (Aber Instruments Ltd) and 58mm diameter<br />

disposable “Coupon” probes. The probes were welded into the RM<br />

Cultibags by Sartorius Stedim. The tests performed with SF9 cells at GSK<br />

used a combination of the Biomass Monitor 220 (Aber Instruments, UK)<br />

and a Futura MRF Mini-Remote Futura pre-amplifier. The media used was<br />

Excell 420 from SAFC Biosciences.<br />

Tests with Rocking Motion bags: One of the challenges of installing<br />

any on-line probe in a rocking motion bioreactor is that the sensor will<br />

be exposed to varying levels of fluid. When the bag is used at the<br />

minimum recommended volume, the probe will also be exposed<br />

momentarily to the gaseous headspace of the bioreactor. In the very first<br />

tests with the disposable probe installed in a 5L Sartorius Stedim Biotech<br />

CultiBag RM filled with just media, the unfiltered capacitance and<br />

conductivity profiles were shown to become increasingly noisier as the<br />

speed of the rocking action was increased.<br />

An advanced rocker filter algorithm, including an “anti-beat” mechanism,<br />

was developed to work automatically at varying rocker speeds such that<br />

the Futura does not have to monitor the rocking speed or synchronise<br />

the readings to the angle of the rocking table. Further experiments were<br />

then performed with disposable probes mounted within a simulated bag<br />

platform on a rocker set at a higher than average speed for cell culture<br />

applications (47rpm). The volume of media (500ml) was set to a<br />

maximum depth of 20mm at full tilt and the minimum depth over the<br />

electrodes was 1mm during the rocking process.<br />

The resulting capacitance and conductivity traces showed that a normal<br />

average filter flattened out the noise, however, if only the averaging filter is<br />

used, the output filtered value was markedly lower than the steady state<br />

value. The new rocker algorithm successfully filtered out the noise caused by<br />

the rocking motion and was far better at maintaining the steady state value.<br />

The disposable probe also has been assessed with a prototype 50-L<br />

Sartorius Stedim Biotech CultiBag RM (25L working volume) in monitoring<br />

thegrowthofSf9insectcells(Figure1).Theprobetrackedviablecell<br />

density before addition of a baculovirus for transient recombinant protein<br />

expression. The biomass probe successfully detected a valid infection by<br />

showing a rapid increase in signal caused by increasing volumes of the<br />

infected cells.<br />

Conclusions: Use of RF impedance to monitor cell culture processes is<br />

well established in biopharmaceutical applications. Introduction of the<br />

Futura biomass monitor allows this technology to be used with<br />

confidence on rocking motion disposable bioreactors, from process<br />

development through cGMP production.<br />

Acknowledgements<br />

Robert Tanner of GSK, UK, and Henry Weichert of Sartorius Stedim,<br />

Germany, are gratefully acknowledged for providing data and<br />

photographs for this poster.<br />

Reference<br />

1. Carvell JP, Williams J, Lee M, Logan D: On-Line Monitoring of the Live Cell<br />

Concentration in Disposable Bioreactors (poster). European Society for<br />

Animal Cell Technology biennial conference, Dublin, Ireland 2009.<br />

P126<br />

Optimization of HEK 293 cell growth by addition of non-animal derived<br />

components using design of experiments<br />

Laura Cervera, Sonia Gutiérrez, Francesc Gòdia, María M Segura *<br />

Departament d’Enginyeria Química, Universitat Autònoma de Barcelona,<br />

Bellaterra, Barcelona, 08193, Spain<br />

E-mail: mersegura@gmail.com<br />

BMC Proceedings 2011, 5(Suppl 8):P126<br />

Page 169 of 181<br />

Background: Mammalian cells are a widely used expression platform for<br />

the production of recombinant therapeutic proteins or viral particle-based<br />

vaccines since they typically perform appropriate protein posttranslational<br />

modifications and authentic viral particle assembly. Of the<br />

available mammalian cells, HEK 293 is one of the most industrially<br />

relevant cell lines because it is cGMP compliant and is able to grow in<br />

suspension in a variety of commercial serum-free media. Of note,<br />

production of human therapeutics in mammalian cell culture has become<br />

more and more stringent in past recent years and not only demands<br />

serum-free but also animal-component free production conditions to<br />

ensure safety. This work is part of a project aimed to optimize HEK 293<br />

cell growth by addition of non-animal derived components to serum-free<br />

and protein-free media through design of experiments (DoE) in order to<br />

maximize productivity of a recombinant VLP vaccine by PEI-mediated<br />

transient transfection.<br />

Materials and methods: Thecelllineusedinthisworkisaserum-free<br />

suspension-adapted HEK 293 cell line from a cGMP master cell bank from<br />

the Biotechnology Institute of the National Research Council in Montreal,<br />

Canada. Cells were maintained in exponential growth in 125-mL<br />

disposable polycarbonate Erlenmeyer flasks shaken at 110 rpm using an<br />

orbitalshakerplacedinanincubator with a 37°C, humidified, 5% CO 2<br />

atmosphere. Cell count and viability were determined using trypan blue<br />

and a microscope counting chamber.<br />

Results: We have analyzed the kinetics of HEK 293 cell growth in HyQ<br />

SFM4 Transfx293 from HyClone Thermo Scientific (Logan, UT, USA).<br />

The cells grow to a maximum concentration of ~ 3×10 6 cells/ml with<br />

over 90% viability and show an average doubling time of 24 h. In<br />

addition, HEK 293 cell growth was assessed in two other commercial<br />

serum-free culture media, namely ExCell 293 from SAFC Biosciences<br />

(Hampshire, UK) and Freestyle 293 from Invitrogen (Carlsbad, CA,<br />

USA) both compatible with PEI-mediated transient transfection ,<br />

showing similar results (Figure 1a). The effect of foetal bovine sera<br />

(FBS) in these serum-free culture media was also evaluated. Cells can<br />

triplicate their maximum cell densities in the presence of FBS (i.e.<br />

they reach 9,3×10 6 cells/ml in HyQ SFM4 Transfx293 medium + 10%<br />

FBS).<br />

Due to the important effect of serum on HEK 293 cell growth, we decided<br />

to evaluate the effect of non-animal derived serum components on cell<br />

growth in attempt to improve cell densities while keeping animal-origin<br />

free production conditions. For these studies, a pre-defined mixture of<br />

supplements composed of r-albumin (1 g/L), r-insulin (10 mg/L), rtransferrin<br />

(10 mg/L) from Merck Millipore (Kankakee, IL, USA), and an inhouse<br />

developed animal-component free lipid mix (1X) composed of<br />

synthetic cholesterol (SyntheChol®, Sigma-Aldrich, Steinheim, Germany),<br />

fatty acids (F7050, SAFC Biosciences), tocopherol (T1157, Sigma) and<br />

emulsifying agents (PS80, Sigma) at concentrations recommended in the<br />

literature [1] were used. HEK 293 cell density is improved in the presence<br />

of the mix, but only in Freestyle medium a significant difference is<br />

observed (Figure 1b). This medium was selected for further optimization<br />

by DoE.<br />

Screening of supplements with significant effect on HEK 293 cell growth<br />

was performed using a Placket-Burman experimental design [2]. Two<br />

levels of concentrations were assigned to each variable: a low one with<br />

no additives and a high one based on the typically recommended values<br />

mentioned above. Using this strategy, we were able to determine in 12<br />

experimental runs (performed in duplicate) that r-insulin, r-transferrin and<br />

an in-house developed lipid mix positively affect HEK 293 cell growth in<br />

serum-free media formulations, whereas r-albumin showed no significant<br />

effect (Figure 1c).<br />

Optimal concentrations for each supplement showing a significant effect<br />

on HEK 293 cell growth were defined based on a Box-Behnken<br />

experimental design [3]. Three levels of concentrations for each variable<br />

were selected (Table 1). Using this experimental design, we were able to<br />

define in 15 experimental runs (performed in duplicate) a model that<br />

accurately predicts HEK 293 cell concentrations in the presence of<br />

different concentrations of r-insulin (r-Ins), r-transferrin (r-Trans) and lipid<br />

mix (LipMix) (Table 1).<br />

The Box-Behnken model equation was:<br />

Cell density (10 6 cells/mL) = 4,53 + 0,63 × r-Ins - 0,17 × r-Trans - 0,41 ×<br />

LipMix - 0,68 × r-Ins × r-Trans + 0,02 × r-Ins × LipMix - 0,07 × rTrans ×<br />

LipMix - 0,18 × r-Ins 2 - 0,45 × r-Trans 2 - 1,02 × LipMix 2


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Page 170 of 181<br />

Figure 1(abstract P126) Optimization of HEK 293 cell growth a) The effect of 10% FBS supplementation in commercially av ailable serum-free media,<br />

b) Growth kinetics of HEK 293 cells in 3 different serum-free protein-free formulations (bI, bII and bIII) in the presence or absence of a pre-defined mixture of<br />

supplements, c) Effect of recombinant proteins or synthetic lipid mix on HEK 293 cell growth, d) Response surface graphs. HEK 293 peak cell density as a<br />

function of the concentrations of Lipid Mix (X) vs. r-Transferrin (mg/L) (dI), r-Transferrin (mg/L) vs. r-Insulin (mg/L) (dII) and Lipid Mix (X) vs. r-Insulin (mg/L)<br />

(dIII) based on Box-Behnken experimental results. Cell density values presented are in millions of cells/mL and represent mean ± SD (n=3).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P126) Box-Behnken results. a Three levels of concentrations for each variable including a maximum<br />

(1), a minimum (-1) and a center point (0) were used. Values shown in parenthesis are concentrations employed for<br />

r-transferrin and r-insulin (mg/mL) and lipid mix (based on Synthechol® concentrations provided in X). b Cell density<br />

values presented are mean of duplicate runs in million cells/mL<br />

EXP N° r-Insulin a<br />

r-Transferrin a<br />

Lipid Mix a<br />

Max Cell density b<br />

Experimental According to model<br />

1 -1 (1) -1 (1) 0 (1) 3,1 2,8<br />

2 1 (20) -1 (1) 0 (1) 5,5 5,4<br />

3 -1(1) 1 (20) 0 (1) 3,7 3,8<br />

4 1 (20) 1 (20) 0 (1) 3,3 3,7<br />

5 -1 (1) 0 (10) -1 (0.1) 3,1 3,1<br />

6 1 (20) 0 (10) -1 (0.1) 4,6 4,4<br />

7 -1 (1) 0 (10) 1 (2) 2,0 2,3<br />

8 1 (20) 0 (10) 1 (2) 3,6 3,6<br />

9 0 (10) -1 (1) -1 (0.1) 3,2 3,6<br />

10 0 (10) 1 (20) -1 (0.1) 3,5 3,4<br />

11 0 (10) -1 (1) 1 (2) 2,8 2,9<br />

12 0 (10) 1 (20) 1 (2) 2,8 2,4<br />

13 0 (10) 0 (10) 0 (1) 4,9 4,5<br />

13 0 (10) 0 (10) 0 (1) 4,9 4,5<br />

13 0 (10) 0 (10) 0 (1) 3,8 4,5<br />

Optimal concentrations for each supplement in HEK 293 cell culture<br />

medium were defined based on this model to be 19,8 mg/L of r-insulin,<br />

1,6 mg/L of r-transferrin and 0,9X for the lipid mix. These results can be<br />

inferred from response surface graphs (Figure 1d). Finally, the model was<br />

validated experimentally. For this purpose, the growth kinetics of HEK 293<br />

cells in the optimized cell culture medium was analyzed. In the presence<br />

of the suitable combination/concentrations of supplements, HEK 293 cells<br />

reached a maximum cell density of 5,4x10 6 cells/mL (n=3), same value as<br />

predicted using the Box-Behnken model, as opposed to the<br />

unsupplemented Freestyle medium that supported cell growth up 3x10 6<br />

cells/mL (n=3) (Figure 1a & 1bIII).<br />

Conclusions: Results have shown that by adding a mixture of animal-free<br />

supplements to serum-free culture medium, it is possible to reach high<br />

cell densities comparable to those attained in the presence of FBS while<br />

avoiding the problems derived from its use.<br />

Acknowledgements<br />

We would like to thank Dr. Amine Kamen (BRI-NRC, Canada) for kindly<br />

providing the HEK 293 cell line. The recombinant albumin was a<br />

generous gift from Merck Millipore.<br />

References<br />

1. Keenan J, Pearson D, Clynes M: The role of recombinant proteins in the<br />

development of serum-free media. Cytotechnology 2006, 50:49-56.<br />

2. Plackett RL, Burman JP: The design of optimum multifactorial<br />

experiments. Biometrika 1946, 33:305-325.<br />

3. Box GEP, Behnken DW: Some new three level designs for the study of<br />

quantitative variables. Technometrics 1960, 2:455-475.<br />

P127<br />

Evaluation of three commercial kits for mycoplasma NAT assays:<br />

selection and quality improvement<br />

Fabien Dorange * , Frédérick Le Goff, Nicolas Dumey<br />

Texcell, Evry, France, 91058<br />

E-mail: dorange@texcell.fr<br />

BMC Proceedings 2011, 5(Suppl 8):P127<br />

Background: Mycoplasma testing on cell lines or biological products<br />

used to be performed based on classical methods such as agar and broth<br />

medium and/or indicator cell culture. However, these methods require a<br />

long incubation period and are not adapted for samples, like liveattenuated<br />

vaccine viruses (which can not completely be neutralized) or<br />

Page 171 of 181<br />

cell therapy products (with short shelf life). NAT assays have several<br />

advantages including rapid-time to results, robustness and sensitivity. The<br />

European Pharmacopoeia updated the 2.6.7 section by adding the<br />

detection of Mycoplasma with NAT methods as an alternative to one or<br />

both classical methods.<br />

Texcell’s offers for Mycoplasma testing, which already included<br />

classical methods, were incremended with NAT assay. For this<br />

purpose, 3 commercial kits based on NAT assay were evaluated based<br />

of their claim to meet the European Pharmacopeia guidance for<br />

nucleic acid amplification techniques for Mycoplasma testing,<br />

including sensitivity and range of detection: CytoCheck® from Greiner,<br />

MycoTOOL® from Roche, MycoSEQ® Mycoplasma detection kit from<br />

Life Technologies.<br />

Material and methods: 5 mycoplasma species were chosen among the<br />

9 strains listed in the European Pharmacopeia.<br />

Mycoplasma Pneumoniae: CIP 103766T<br />

Mycoplasma Hyorhinis: ATCC 179891<br />

Acholeplasma Laidlawii: ATCC 23206<br />

Mycoplasma Orale: provided by Greiner Bio-One<br />

Mycoplasma Synoviae: provided by Greiner Bio-One<br />

1 ml of different concentrations of mycoplasma were tested according to<br />

the supplier’s instructions.<br />

Results: For the mycotool results, none of the mycoplasma species tested<br />

were detected above the required limit of detection of 10 cfu/ml.<br />

Investigations showed that quantity of nucleic acids in our experimental<br />

design was too small to be efficiently recovered with the precipitation<br />

based extraction procedure. Indeed, the mycoTOOL® kit was designed to<br />

be used in conjonction with CHO cells (5x10 6 cells/ml), acting like carrier<br />

DNA for the precipitation step. Since this study, the kit’s supplier has<br />

included a carrier DNA for samples containing low level of nucleic acid.<br />

Both remaining selected kits (MycoSEQ® and CytoCheck®) met the<br />

sensitivity parameters and were compared (Table 1).<br />

Although these kits use a different technology, similar results (sensitivity,<br />

range of detection) were obtained. The lower sensitivity observed with<br />

the MycoSEQ® kit for M. Pneumoniae is explained by the presence of nonviable<br />

mycoplasmas induced during thawing/freezing cycle for stocks<br />

preparation.<br />

According to its safer use (no post-amplification handling), its lower cost<br />

and quantitation possibilities, the MycoSEQ® kit was preferred.<br />

Performance validation was conducted in Texcell facilities using the<br />

MycoSEQ® Mycoplasma detection kit.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P127) Comparison of the MycoSEQ® Mycoplasma detection kit and CytoCheck® kit<br />

KIT supplier<br />

Parameters MycoSEQ® CytoCheck® Priority<br />

Applied Biosystems Greiner<br />

tested sensitivity 40% saturation by<br />

pulsing or constant aeration with sterile air. A biphasic process scheme<br />

was applied; cell growth phase (0 – 120hcultivationtime)andvirus<br />

propagation phase (120 – 192 h cultivation time). For infection a media<br />

change was performed: cells attached to the microcarriers were allowed<br />

to settle down by stopping agitation for 2 h, after two rinsing steps with<br />

PBS the cell culture medium was completely exchanged against the same<br />

MEM with 1% L-Glutamin but without FBS (infection medium).<br />

Afterwards, a small amount of infection medium containing porcine<br />

Influenza A virus/H1N1/strain 2617 originating from WSV (IDT Biologika<br />

GmbH) with a moi of 10 -4 and a trypsine concentration of 40 units/ml<br />

was given into the bioreactor. Samples were taken in distinct intervals<br />

and aliquoted for offline analytics including cell count with CASY model<br />

TT (Roche Diagnostics), further cell analytics for apoptosis and necrosis<br />

with a flow cytometer Guava easycyte and respective commercial assays<br />

(Millipore GmbH), automated enzymatic assays for quantification of<br />

glucose, lactate, glutamine and ammonia levels (YSI 7100 MBS, Yellow<br />

Springs Instruments) and quantification of porcine Influenza virus with<br />

hemagglutination (HA) and virus titration assay (TCID 50) according to IDT<br />

internal protocols. Online data, especially for temperature, stirrer speed,<br />

pH-value, pO2 and gassing rate were recorded by the software MFCS/DA<br />

(Sartorius Stedim Biotech GmbH).<br />

Results and discussion: After seeding the cells attached rapidly onto the<br />

microcarriers; at first analytics after 24 h cells showed a good distribution<br />

on the microcarriers with typical morphology. Cell number increased up<br />

to a maximum of 2.1 x 10 6 cells/ml with a viability of 92% after 120 h,<br />

which was quite in accordance with expectations when using a carrier<br />

concentrationof2g/linabatchmodeprocess(figure1).Thegrowth<br />

rate µ max in the interval from 24-120 h was 0.037 h -1 which corresponds<br />

to a doubling time of about 19 h. The handling of medium change at the<br />

beginning of the virus propagation phase was tricky but can be easily<br />

improved by using customized bags for microcarrier cultivations with<br />

media changes. The cell culture during growth phase (both populations,<br />

cells on microcarriers and in suspension) was also monitored via nexin<br />

assay determining the part of apoptotic (early and late phase) and<br />

necrotic cells. The obtained data confirm the cell count and viability data<br />

measured with the CASY TT system and microscopic observations.<br />

The viral infection proceeded very rapidly with first signs of Influenza<br />

related cytopathic effect 24 h post infection, leading to virus yields of<br />

maximum log 3.0 HA units and 10 8.00 TCID50/ml after 48 h post infection.<br />

Afterwards the infectious titer dropped slightly while HA remained stable.<br />

The virus yields achieved were 2-4-fold higher compared to standard


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P128) MDBK cell growth and viability for cell populations on microcarriers and in suspension during cultivation in a 50 l scale<br />

disposable stirred-tank bioreactor run for propagation of porcine Influenza A/H1N1 (black line indicates virus infection).<br />

roller bottle cultivations. For a non optimized process, these virus yields<br />

are very promising. Very likely, by finetuning of infection parameters<br />

(moi, trypsine concentration), harvest time point and increase of cell<br />

numbers before infection even higher titers are achievable.<br />

One of the most important advantages of a bioreactor system is its<br />

controlled surrounding in terms of pH and oxygen level. In this study that<br />

fact was proven very well by the good performance of the control system<br />

(figure 2 and 3). The profile for pO2 and air flow rate corresponds well to<br />

the cultivation curve; in the first hours of cultivation the pulsing aeration<br />

can be seen as only a few cells were in the reactor system. With<br />

increasing cell numbers a constant air flow is applied starting approx.<br />

after 48 h cultivation time leading to a maximal airflow of 3.5 l/min<br />

keeping the oxygen level constant also when higher cell concentrations<br />

are present. The virus infection led to a rapid decrease in oxygen uptake;<br />

Page 173 of 181<br />

at the end of cultivation oxygen consumption tended to be zero (figure<br />

2). Temperature and stirrer speed control was not a problem throughout<br />

cultivation (figure 3); also control of pH value worked very well (7.40 ±<br />

0.15).<br />

Conclusions: The results of this study prove a successful tech transfer for<br />

a porcine Influenza virus production process from roller bottles into a<br />

novel disposable STR bioreactor system with advances in both cell and<br />

virus yields and process control possibilities. These are important factors<br />

in near future, keeping upstream processing competitive in terms of<br />

prices, productivity and surely also from a regulatory point of view. It is a<br />

further step in order to be prepared for pandemic situations as it was<br />

seen for 2009 H1N1 occurrence. There should be no obstacles for<br />

implementing such systems into GMP surrounding. As result of the study<br />

also a lot of valuable data were generated which can be used for process<br />

Figure 2(abstract P128) Online data from cultivation in a 50 l scale disposable stirred-tank bioreactor run for propagation of porcine Influenza A/H1N1<br />

for oxygen partial pressure and air flow rate.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 3(abstract P128) Online data from cultivation in a 50 l scale disposable stirred-tank bioreactor run for propagation of porcine Influenza A/H1N1<br />

for temperature, stirrer speed and pH-value.<br />

validation and establishment of descriptive mathematical process models<br />

thus developing a deeper understanding of production processes for<br />

biologicals.<br />

P129<br />

Effect of influenza virus infection on key metabolic enzyme activities in<br />

MDCK cells<br />

Robert Janke 1* , Yvonne Genzel 1 , Maria Wetzel 2 , Udo Reichl 1,2<br />

1 Max Planck Institute for Dynamics of Complex Technical Systems,<br />

Bioprocess Engineering group, Sandtorstraße 1, 39106 Magdeburg, Germany;<br />

2 Chair for Bioprocess Engineering, Otto von Guericke University of<br />

Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany<br />

E-mail: janke@mpi-magdeburg.mpg.de<br />

BMC Proceedings 2011, 5(Suppl 8):P129<br />

Background: Influenza, or “flu”, is an upper respiratory tract infection<br />

caused by a virus belonging to the family of Orthomyxoviridae. Influenza<br />

can pose a serious risk to the health of mainly the elderly, the very<br />

young, and to people suffering from medical conditions (e.g. weak<br />

immune system). For example, seasonal influenza strains are fatal to<br />

more than 50,000 people annually in the United States alone [1]. The<br />

most effective measure for preventing influenza-related morbidity and<br />

mortality is annual vaccination. Seasonal influenza vaccines are almost<br />

exclusively produced using the traditional egg-based manufacturing<br />

process. However, the main limitation of egg-based technology<br />

(especially in the case of a pandemic) is the time-consuming production<br />

process (~6 months). Furthermore, people with serious egg allergy<br />

cannot be vaccinated when trace amounts of egg protein remain in the<br />

final formulation. Therefore, new production processes using continuous<br />

cell lines for influenza vaccine manufacturing are currently being<br />

established [2].<br />

Influenza viruses take advantage of the host cell metabolism to replicate<br />

their genetic material and to synthesize viral proteins. The influenza virus<br />

particle consists of three major parts: the ribonucleocapsid, the matrix<br />

protein M1, and the envelope, which is derived from the plasma<br />

membrane of the host cell. The lipid bilayer contains the ion channel<br />

protein M2 and the immunogenic glycoproteins hemagglutinin and<br />

neuraminidase [3]. The replication cycle of influenza viruses including<br />

entry, uncoating, genome transcription and replication, assembly and<br />

release has been studied extensively with type A strains [4]. So far, only<br />

few studies have characterized the influence of influenza infection on the<br />

Page 174 of 181<br />

central carbon metabolism of host cells [5]. Madin-Darby canine kidney<br />

(MDCK) cells are considered a suitable substrate for cell culture-based<br />

influenza vaccine manufacturing [2,6]. In this study, key metabolic<br />

enzyme activities were analyzed in MDCK cells infected with an influenza<br />

A virus strain to improve our understanding of virus-host cell interaction<br />

and cell response.<br />

Materials and methods: All chemicals and enzymes were purchased<br />

from Sigma (Taufkirchen, Germany) or Roche (Mannheim, Germany).<br />

Adherent MDCK cells obtained from the ECACC (No. 84121903) were<br />

routinely cultured in 6-well plates containing 4 mL of GMEM-based<br />

medium (2 mM glutamine, 30 mM glucose, 10% (v/v) fetal calf serum,<br />

2 g/L peptone, 48 mM NaHCO3) inaCO2 incubator at 37 °C and 5% CO2<br />

to the stationary phase (~5 days of growth, 4.0-4.2 x 10 6 cells) [7]. Cell<br />

concentration and viability was determined for samples from 6-well<br />

plates as described previously [8]. MDCK cells were either mock-infected<br />

or infected with MDCK cell-adapted human influenza virus A/Puerto Rico/<br />

8/34 (H1N1) from the Robert Koch Institute (Berlin, Germany) at a<br />

multiplicity of infection of 20 as described previously [5,9]. Cells were<br />

washed twice with ice-cold phosphate-buffered saline 9 hours post<br />

infection (hpi), and the complete plate was then snap-frozen in liquid<br />

nitrogen and stored at -80 °C until further use. After thawing, samples<br />

were extracted by sonification on maximum power for 30 s with 1 mL<br />

extraction buffer [7] and kept at 0–4 °C. To remove cell debris, samples<br />

were centrifuged at 16,000 x g for 5 min. The supernatant was used to<br />

measure the respective enzyme activities. The procedure for the enzyme<br />

activity analysis and the details for the specific assay mixes were as<br />

described previously [7].<br />

Results: The maximum catalytic activities of 28 enzymes from central<br />

carbon metabolism were measured under substrate saturation using a<br />

recently developed assay platform for mammalian cells [7]. Table 1 shows<br />

the maximum metabolic enzyme activities in mock-infected and influenza<br />

A (H1N1) infected MDCK cells. The activities of different enzymes<br />

comprise several orders of magnitude. The overall range covers values<br />

from 0.24±0.10 nmol/min/10 6 cells (pyruvate dehydrogenase, PDH) to<br />

10348.06±1663.65 nmol/min/10 6 cells (triose-phosphate isomerase, TPI) in<br />

H1N1 and mock-infected cells, respectively. Highest activities (>1000<br />

nmol/min/10 6 cells) were found for TPI, pyruvate kinase, lactate<br />

dehydrogenase, and malate dehydrogenase, while the other activities<br />

were in the range of 1 to 500 nmol/min/10 6 cells. Very low enzyme<br />

activities, which indicate possible rate-limiting steps in the respective<br />

metabolic pathway, were found for PDH, pyruvate carboxylase (PC),<br />

NAD + -dependent isocitrate dehydrogenase (NAD-ICDH), and glutamine


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Table 1(abstract P129) Maximum enzyme activities of glycolysis, pentose phosphate pathway, TCA cycle, and<br />

glutaminolysis in MDCK cells infected with influenza A (H1N1) compared to mock-infected cells<br />

Enzyme activities in adherent MDCK cells a (nmol/min/10 6 cells)<br />

Enzyme EC number Mock-infected H1N1 infected<br />

Glycolysis<br />

Hexokinase 2.7.1.1 21.80 ± 3.95 21.89 ± 1.48<br />

Phosphoglucose isomerase 5.3.1.9 465.12 ± 73.94 456.93 ± 111.90<br />

Phosphofructokinase 2.7.1.11 29.28 ± 6.14 30.56 ± 2.08<br />

Fructose-1,6-bisphosphate aldolase 4.1.2.13 23.62 ± 3.32 20.74 ± 8.41<br />

Triose-phosphate isomerase 5.3.1.1 10348.06 ± 1663.65 10148.62 ± 698.01<br />

Glyceraldehyde-3-phosphate dehydrogenase 1.2.1.12 412.90 ± 73.27 413.61 ± 31.61<br />

Pyruvate kinase 2.7.1.40 1004.26 ± 43.53 1001.92 ± 112.95<br />

Lactate dehydrogenase<br />

Pentose phosphate pathway<br />

1.1.1.27 1266.97 ± 134.69 1302.10 ± 107.37<br />

Glucose-6-phosphate dehydrogenase 1.1.1.49 51.91 ± 1.10 62.71 ± 4.59 b<br />

6-phosphogluconate dehydrogenase 1.1.1.44 30.61 ± 5.03 38.13 ± 1.05 b<br />

Transketolase 2.2.1.1 18.63 ± 6.65 19.19 ± 2.52<br />

Transaldolase<br />

Tricarboxylic acid cycle<br />

2.2.1.2 23.11 ± 5.84 23.23 ± 6.30<br />

Pyruvate dehydrogenase 1.2.4.1 0.30 ± 0.07 0.24 ± 0.10<br />

Pyruvate carboxylase 6.4.1.1 0.48 ± 0.11 0.82 ± 0.23 b<br />

Citrate synthase 2.3.3.1 21.55 ± 1.70 25.40 ± 2.84 b<br />

Citrate lyase 2.3.3.8 4.48 ± 0.55 6.00 ± 0.98 b<br />

NAD + -linked isocitrate dehydrogenase 1.1.1.41 0.27 ± 0.02 0.34 ± 0.05 b<br />

NADP + -linked isocitrate dehydrogenase 1.1.1.42 44.27 ± 2.73 44.57 ± 5.37<br />

Fumarase 4.2.1.2 74.62 ± 6.56 77.85 ± 11.75<br />

Malate dehydrogenase<br />

Glutaminolysis<br />

1.1.1.37 1115.42 ± 90.90 1158.66 ± 145.10<br />

Glutaminase 3.5.1.2 3.29 ± 0.29 3.90 ± 0.34 b<br />

Glutamine synthetase 6.3.1.2 0.68 ± 0.25 0.80 ± 0.25<br />

Glutamate dehydrogenase 1.4.1.2 2.54 ± 0.51 2.20 ± 0.31<br />

Alanine transaminase 2.6.1.2 3.21 ± 0.50 3.31 ± 0.03<br />

Aspartate transaminase 2.6.1.1 78.67 ± 5.36 79.12 ± 13.17<br />

Malic enzyme 1.1.1.40 7.61 ± 0.61 9.87 ± 0.62 b<br />

Phosphoenolpyruvate carboxykinase<br />

Miscellaneous<br />

4.1.1.32 138.64 ± 15.33 131.48 ± 11.42<br />

Acetate-CoA ligase 6.2.1.1 2.01 ± 0.13 2.69 ± 0.34 b<br />

a<br />

Mean values and 95 % confidence intervals for three biological replicates.<br />

b<br />

Significantly different from mock-infected cells as determined by Student’s t-test (p ≤ 0.05).<br />

synthetase (


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

Figure 1(abstract P129) Reaction network scheme of the central carbon metabolism of adherent MDCK cells. Enzymes up-regulated in influenza A<br />

(H1N1) infected cells are highlighted in blue. Abbreviations: 6PGDH, 6-phosphogluconate dehydrogenase; ACoAL, acetate-CoA ligase; CL, citrate lyase; CS,<br />

citrate synthase; G6PDH, glucose-6-phosphate dehydrogenase; GLNase, glutaminase; MDH, malate dehydrogenase; ME, malic enzyme; PDH, pyruvate<br />

dehydrogenase; PC, pyruvate carboxylase.<br />

Ltd: Mahy BWJ, ter Meulen V , 10 2010, 3 and 4:634-698[http://onlinelibrary.<br />

wiley.com/doi/10.1002/9780470688618.taw0238/abstract].<br />

5. Ritter JB, Wahl AS, Freund S, Genzel Y, Reichl U: Metabolic effects of<br />

influenza virus infection in cultured animal cells: Intra- and extracellular<br />

metabolite profiling. Bmc Syst Biol 2010, 4.<br />

6. Doroshenko A, Halperin SA: Trivalent MDCK cell culture-derived influenza<br />

vaccine Optaflu (Novartis Vaccines). Expert Rev Vaccines 2009, 8(6):679-688.<br />

7. Janke R, Genzel Y, Wahl A, Reichl U: Measurement of Key Metabolic<br />

Enzyme Activities in Mammalian Cells Using Rapid and Sensitive<br />

Microplate-Based Assays. Biotechnol Bioeng 2010, 107(3):566-581.<br />

8. Genzel Y, Ritter JB, König S, Alt R, Reichl U: Substitution of glutamine by<br />

pyruvate to reduce ammonia formation and growth inhibition of<br />

mammalian cells. Biotechnol Progr 2005, 21(1):58-69[http://onlinelibrary.<br />

wiley.com/doi/10.1021/bp049827d/abstract].<br />

9. Genzel Y, Behrendt I, König S, Sann H, Reichl U: Metabolism of MDCK<br />

cells during cell growth and influenza virus production in large-scale<br />

microcarrier culture. Vaccine 2004, 22(17-18):2202-2208.<br />

P130<br />

Novel human partner cell line for immortalisation of rare antigenspecific<br />

B cells in mAb development<br />

Galina Kaseko * , Marjorie Liu, Qiong Li, Tohsak Mahaworasilpa<br />

The Stephen Sanig Research Institute, Suite G17, National Innovation Centre,<br />

Australian Technology Park, 4 Cornwallis Street, Eveleigh, NSW, 2015,<br />

Australia<br />

E-mail: g.kaseko@ssri.org.au<br />

BMC Proceedings 2011, 5(Suppl 8):P130<br />

It is well-documented that post-translational modification (PTM) events,<br />

such as glycosylation, play an important role in antibody-dependent cellmediated<br />

cytotoxicity (ADCC) [1,2]. In current technological processes,<br />

monoclonal antibody (mAb) production is widely achieved using<br />

heterologous hybridoma systems or genetic engineering using various<br />

Page 176 of 181<br />

non-human cell lines as expression host. As a consequence, PTMs<br />

generated from non-human cell lines may differ from their human<br />

counterparts, resulting in diminished antibody efficacy, aberrant folding<br />

and adverse immunogenic response. Therefore, the use of human partner<br />

cell lines to generate “fully human” mAbs is beneficial as it circumvents<br />

functional complications associated with non-human cell lines.<br />

A human cross-lineage hybrid cell line was developed in our laboratory as<br />

a candidate partner for immortalisation of rare primary human antigenspecific<br />

B lymphocytes using binary electrical cell hybridisation technique<br />

which has been developed in house. This novel partner cell line is a trihybrid<br />

of IL-4 secreting Th2 lymphoblast derived from a patient with<br />

acute lymphoblastic leukaemia (T), CD20 + B lymphoblast, also derived<br />

from a patient with acute lymphoblastic leukaemia (W), and IL-6 secreting<br />

peripheral blood-derived CD14 + monocyte (M). The selection of cell<br />

phenotypes used to create the tri-hybrid was based on factors known to<br />

maintain and promote antibody production.<br />

The resulting tri-hybrid (WTM) displayed characteristics of mixed CD<br />

phenotypes with the majority of cells being CD20 + (95%) with coexpression<br />

of CD4 + (54%) and CD14 + (24%). It secreted IL-4, IL-6, IL-8 and<br />

GM-CSF but was negative for IL-1A, IL-1B, IL-2, IL-5, IL-10, IL-12, IL-13 and<br />

IL-17.Thecelllinedidnotexpressthe tumour suppressor protein, p53,<br />

and neither did it secrete immunoglobulins (Ig)/ Ig chains nor were they<br />

expressed on the surface.<br />

Table 1(abstract P130) Success rate of stable hybrid<br />

generation and Ig producing hybrids<br />

Event Success rate,<br />

Hybridisation 100 (%) of attempts<br />

Stable hybrids 48 (%) of hybridised<br />

Ig producing hybrids 23 (%) of stable hybrids


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

WTM cells were then used as a fusion partner with primary antigenexperienced<br />

CD19 + B cells which had been isolated from peripheral<br />

blood, activated in vitro, and the resultant hybrids were sorted for IgM +<br />

and IgG + expression. 100% hybridisation success rate was achieved using<br />

a binary electrical cell hybridisation technique and the number of<br />

resulting stable hybrids varied from 48% to 78% depending on the<br />

phenotype of B lymphocytes used in experiments. 23% to 68% of those<br />

stable hybrids secreted Ig with production ranging between 0.2 to 1.2 ìg/<br />

10 6 cells (Table 1). Cytokine screening of some of the Ig producing<br />

hybrids revealed a cytokine profile which was inherently different to that<br />

of the WTM partner cell line. The Ig producing hybrids concurrently<br />

expressed IL-10 and GM-CSF but not IL-4, IL-6 or IL-8. These hybrids were<br />

also positive for TGF-â, RANTES, MIP, MCP and MDC.<br />

In conclusion, major advantages of our method involve the rapid<br />

generation of stable Ig producing hybrids from a small B lymphocyte<br />

population size (50 cells) and the elimination of conventional laborious<br />

screening methods for hybrids and Ig producing clones. Thus, when the<br />

number of rare antigen-specific B cells available is a limiting factor in<br />

generating hybridoma, EBV transfection or direct sequencing, binary<br />

electrical B lymphocyte hybridisation with WTM cells can provide a very<br />

attractive approach for the generation of stable hybrid cell lines<br />

producing monoclonal antibodies.<br />

References<br />

1. Raju TS: Impact of Fc Glycosylation on Monoclonal Antibody Effector<br />

Functions and Degradation by Proteases. In Current Trends in Monoclonal<br />

Antibody Development and Manufacturing Biotechnology: Pharmaceutical<br />

Aspects NY 3, USA: Springer Science + Business Media: Shire S 2010 1001,<br />

XI(6):249-269.<br />

2. Werner RG, Kopp K, Schlueter M: Glycosylation of therapeutic proteins in<br />

different production systems. Acta Paediatri 2007, 96:17-22.<br />

Page 177 of 181<br />

P131<br />

Application of the novel and convenient IR/MAR gene amplification<br />

technology to the production of recombinant protein pharmaceuticals<br />

Yoshio Araki 1 , Chiemi Noguchi 1 , Tetsuro Hamafuji 2 , Hiroshi Nose 2 ,<br />

Daisuke Miki 3 , Noriaki Shimizu 1*<br />

1 Graduate School of Biosphere Science, Hiroshima University, Higashihiroshima,<br />

739-8521, Japan; 2 Transgenic Inc., Kobe, 650-0047, Japan; 3 Tosoh<br />

Co., Tokyo, 252-1123, Japan<br />

E-mail: shimizu@hiroshima-u.ac.jp<br />

BMC Proceedings 2011, 5(Suppl 8):P131<br />

Amplification of DHFR gene in CHO cells by selection of MTx has been<br />

widely applied to the establishment of stable cell lines that efficiently<br />

produce recombinant protein pharmaceuticals. However, the DHFR/<br />

MTx technology was highly time-and labor-consuming. On the other<br />

hand, we had found that a plasmid bearing a mammalian replication<br />

initiation region (IR) and a matrix attachment region (MAR) initiates<br />

gene amplification in mammalian cells, and it quite efficiently<br />

generate the chromosomal HSR and/or the extrachromosomal DMs<br />

[1,2]. This is a completely original technology of gene amplification,<br />

and we have revealed the mechanism why and how such plasmid<br />

may mimic gene amplification [2,3]. Now, we aimed to adopt this<br />

technology to the industrial production of recombinant protein<br />

pharmaceuticals.<br />

We constructed plasmids with or without IR/MAR, with several promoters<br />

for drug resistant gene, antibody gene or Fc receptor gene, and with<br />

various orientations of these elements. The plasmid DNA with several<br />

physical structures were transfected to human COLO 320DM or hamster<br />

CHO DG44 cells, with or without another DNA. The transformed cells<br />

Figure 1(abstract P131) The IR/MAR plasmid can generates DMs (A), HSR (B), Ladder-HSR (C), and Fine ladder-HSR (D). Among the metaphase<br />

chromosome spread, we detected the plasmid sequence by FISH in green. Gene expression was generally higher from DMs than HSR. However, DMs are<br />

usually hard to be generated in CHO cells. The IR/MAR plasmid can efficiently generate the ladder and the fine ladder structure that is active in<br />

transcription.


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

were selected by various conditions. The polyclonal transformants or the<br />

cloned cells were evaluated by the protein production (ELISA), as well as<br />

by the structure of amplified region (FISH).<br />

As a result, the usage of IR/MAR technology enabled us to obtain cells, in<br />

which the introduced genes were amplified to a few hundreds to<br />

thousands copies per cells as DMs or HSR of various size and shape<br />

(Figure 1), which depended both on the vector constructs and the host<br />

cell lines. Such stable cells with amplified genes could be obtained within<br />

one month, and the protein production was increased more than a<br />

hundred-fold compared with the case without IR/MAR. A cell clone<br />

showed the specific production rate that reached almost the highest<br />

reported for antibody protein (45 pg/cell/day). Furthermore, we have<br />

found several novel ways that further improve the protein production<br />

level. For example, the combination of the IR/MAR and the DHFR/MTx<br />

technologies synergistically work and far more rapidly and easily generate<br />

the cells of higher production rate than previously.<br />

In conclusion, the IR/MAR technology is a novel highly-competitive<br />

technology for use in recombinant protein production, and it further has<br />

potentials for improvement.<br />

References<br />

1. Shimizu N, Miura Y, Sakamoto Y, Tsutsui K: Plasmids with a Mammalian<br />

Replication Origin and a Matrix Attachment Region Initiate the Event<br />

Similar to Gene Amplification. Cancer Res 2001, 61:6987-6990.<br />

2. Shimizu N, Hashizume T, Shingaki K, Kawamoto J: Amplification of<br />

plasmids containing a mammalian replication initiation region is<br />

mediated by controllable conflict between replication and transcription.<br />

Cancer Res 2003, 63:5281-5290.<br />

3. Harada S, Sekiguchi N, Shimizu N: Amplification of a plasmid bearing a<br />

mammalian replication initiation region in chromosomal and<br />

extrachromosomal contexts. Nuc Acids Res 2011, 39:958-969.<br />

P132<br />

About making a CHO production cell line “research-friendly” by genetic<br />

engineering<br />

Bernd Voedisch 1* , Agnès Patoux 1 , Jildou Sterkenburgh 2 , Mirjam Buchs 1 ,<br />

Emily Barry 3 , Cyril Allard 1 , Sabine Geisse 1<br />

1 Novartis Pharma AG, Basel, Switzerland; 2 University of Cambridge,<br />

Cambridge, United Kingdom; 3 Cardiff University, Cardiff, United Kingdom<br />

BMC Proceedings 2011, 5(Suppl 8):P132<br />

The use of Chinese Hamster Ovary (CHO) cells for transient gene expression is<br />

gaining importance steadily, as it has been shown that both quality and<br />

quantity of proteins derived from CHO cells differs from and can even be<br />

superior to material derived from Human Embryonic Kidney (HEK293) cells<br />

[1-4]. In order to augment yields HEK cell lines have been genetically modified,<br />

by e.g. stable integration of an expression cassette coding for the Epstein-Barr<br />

Virus Nuclear Antigen 1 (EBNA1) in conjunction with expression plasmid<br />

vectors containing the EBNA1 interaction site oriP. Here we describe the<br />

generation of a CHO cell line stably expressing the EBNA1 gene to enhance<br />

yields after large scale transient transfection with polyethylenimine (PEI).<br />

An expression plasmid featuring the EBNA1 gene was transfected into an<br />

in-house available CHO wild type cell line by nucleofection and several<br />

stable pools were established by antibiotic selection. In a first approach,<br />

133 clones were then isolated by limiting dilution cloning and<br />

subsequently expanded for further analysis. The approach aimed at<br />

identifying clones showing both the presence of EBNA1 protein and<br />

enhanced yields after PEI-mediated transient expression of a reporter<br />

protein at the same time.<br />

Cell lysates or nuclear and cytoplasmic protein extracts from these clones<br />

were tested for presence of EBNA1 protein by Western Blot. An inlicensed<br />

cell line, CHO EBNALT85 (Icosagen AS, Tartu, Estonia) expressing<br />

the full length EBNA1 gene, and the HEK293-6E cell line (from the group<br />

of Y. Durocher, NRC, Canada) expressing a truncated EBNA1 gene served<br />

as positive controls. A number of commercially available anti-EBNA1<br />

antibodies were tested in Western blotting, but most antibodies failed to<br />

detect the EBNA1 protein produced by these positive control cells. Only<br />

one antibody (Antibody 1EB12, Santa Cruz Biotechnology) was identified<br />

that detected the EBNA1 proteins in both positive control cell lines.<br />

However, by applying this antibody on the 133 generated clones it<br />

became obvious that in most of them the EBNA1 protein appeared to be<br />

largely degraded.<br />

Page 178 of 181<br />

The functionality of the EBNA1 protein in the cells was probed by PEImediated<br />

transient transfection of expression plasmids encoding the<br />

gene for the reporter protein Secreted Alkaline Phosphatase (SeAP). SeAP<br />

expression levels were compared with respect to presence or absence of<br />

the EBNA1 interaction sequence oriP on the SeAP expression plasmid.<br />

Only one clone showed a twofold enhanced production level of the<br />

reporter protein compared to the non-engineered parental CHO cell line.<br />

However, in this specific clone no EBNA1 protein could be detected by<br />

Western Blot, and the enhanced expression level was independent from<br />

the presence or absence of the oriP sequence on the SeAP expression<br />

plasmid. We therefore conclude that this clone does not possess a<br />

functional EBNA1 protein and an EBNA1/oriP based enhancement of<br />

productivity. The mechanism underlying the enhanced yield of the<br />

reporter protein in this particular clone remains currently elusive.<br />

In an alternative approach flow cytometric cell sorting was applied to<br />

generate CHO clones producing a functional EBNA1 protein. Using the<br />

EBNA1 positive CHO cell line CHO EBNALT85 it could be shown that<br />

expressing eGFP from a transiently transfected expression plasmid with<br />

oriP sequence resulted in a 30- to 40-fold increase of highly fluorescent<br />

cells in comparison to an expression plasmid lacking the oriP sequence.<br />

Thus, the already generated CHO pools stably transfected with the EBNA1<br />

gene were transiently supertransfected with an eGFP expression plasmid<br />

containing the oriP sequence. Highly fluorescent cells were collected by<br />

cell sorting and expanded. Analysis by Western Blot revealed that this<br />

sub-poolwasenrichedforcellsexhibiting the presence of a full length<br />

EBNA1 protein besides a major breakdown product. Transient transfection<br />

of this sub-pool with SeAP expression vectors led to the conclusion that<br />

the pool contained cells with a functional EBNA1 protein capable of<br />

enhancing SeAP yields in dependence on the presence of the oriP<br />

sequence on the SeAP expression plasmid. Clones were again generated<br />

from this sub-pool by limiting dilution cloning and characterized in<br />

analogy to the first approach. 9 out of 12 clones that were analyzed<br />

further showed increased levels of reporter protein production in<br />

dependence on the presence of the oriP element on the expression<br />

plasmid. The series of clones exhibiting highest expression levels will now<br />

be further evaluated by expression of other protein candidates.<br />

References<br />

1. Gaudry JP, Arod C, Sauvage C, Busso S, Dupraz P, Pankiewicz R,<br />

Antonsson B: Purification of the extracellular domain of the membrane<br />

protein GlialCAM expressed in HEK and CHO cells and comparison of<br />

the glycosylation. Protein Expr Purif 2008, 58(1):94-102.<br />

2. Haack A, Schmitt C, Poller W, Oldenburg J, Hanfland P, Brackmann HH,<br />

Schwaab R: Analysis of expression kinetics and activity of a new Bdomain<br />

truncated and full-length FVIII protein in three different cell<br />

lines. Ann Hematol 1999, 78(3):111-116.<br />

3. Suen KF, Turner MS, Gao F, Liu B, Althage A, Slavin A, Ou W, Zuo E,<br />

Eckart M, Ogawa T, et al: Transient expression of an IL-23R extracellular<br />

domain Fc fusion protein in CHO versus HEK cells results in improved<br />

plasma exposure. Protein Expr Purif 2010, 71(1):96-102.<br />

4. Van den Nieuwenhof IM, Koistinen H, Easton RL, Koistinen R, Kamarainen M,<br />

Morris HR, Van Die I, Seppala M, Dell A, Van den Eijnden DH: Recombinant<br />

glycodelin carrying the same type of glycan structures as contraceptive<br />

glycodelin-A can be produced in human kidney 293 cells but not in<br />

chinese hamster ovary cells. Eur J Biochem 2000, 267(15):4753-4762.<br />

P133<br />

CAP-T cell expression system: a novel rapid and versatile human cell<br />

expression system for fast and high yield transient protein expression<br />

Jens Wölfel * , Ruth Essers, Corinna Bialek, Sabine Hertel,<br />

Nadine Scholz-Neumann, Gudrun Schiedner<br />

CEVEC Pharmaceuticals GmbH, Cologne, Germany<br />

BMC Proceedings 2011, 5(Suppl 8):P133<br />

Backround: CAP (CEVEC’s Amniocyte Production) cells are an<br />

immortalized cell line based on primary human amniocytes. They were<br />

generated by transfection of these primary cells with a vector containing<br />

the functions E1 and pIX of adenovirus 5. CAP cells allow for competitive<br />

stable production of recombinant proteins with excellent biologic activity<br />

and therapeutic efficacy as a result of authentic human posttranslational<br />

modification. In order to gain access to the benefits of the CAP technology<br />

also for early research, target evaluation or assay development, the


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

transient expression system CAP-T was developed. CAP-T cells are based<br />

on the original CAP cells and additionally express the large T antigen of<br />

simian virus 40 (SV40).<br />

Results: To characterize the CAP-T expression system, they were<br />

transiently transfected with an expression plasmid for the highly complex<br />

and glycosylated human a1-Antitrypsin (hAAT). This resulted in<br />

remarkably high expression levels of up to 60 mg/L. These levels could<br />

be even increased 2.5 fold by adding an SV40 origin of replication<br />

(SV40ori) to the expression vector (Figure 1). When compared to HEK293T<br />

cells, CAP cells showed a significantly higher expression titer than<br />

HEK293T cells (data not shown). In order to understand these<br />

phenomena, the copy number of the expression plasmid, upon<br />

transfection, was determined. CAP-T efficiently replicated the expression<br />

plasmid containing the SV40ori, resulting in increasing copy numbers per<br />

cell over time yielding in about 4 times higher copy numbers than in<br />

HEK293T cells transfected with the same plasmid and in CAP-T cells<br />

transfected with the plasmid lacking the SV40ori (data not shown) and<br />

consequently in higher expression levels. In order to establish a more<br />

scalable transfection method than nucleofection, the suitability of<br />

different transfection reagents for the transfection of CAP-T cells was<br />

determined. In these experiments beside nucleofection, 293fectin and<br />

polyethylenimin (PEI) based transfection methods showed best results in<br />

Page 179 of 181<br />

Figure 1(abstract P133) Transient expression of hAAT in CAP-T: Two plasmids containing a hAAt expression cassette were transfected by nucleofection<br />

(1 x 10 7 cells). The plasmid containing a SV40ori (ori) yielded about 2.5 time higher expression levels at maximum product concentration than a plasmid<br />

lacking this ori (w/o ori).<br />

small scale transfections, enabling the scalability of the transient<br />

transfection in CAP-T cells (data not shown). With the PEI based<br />

transfection method hAAT could be produced also in 300 mL shaking<br />

culture and 1L bioreactor with product titers of up to 180 mg/L in simple<br />

batch processes of up to 10 days (data not shown). Several other<br />

glycosylated proteins have been tested in transient transfections of CAP-T<br />

cells yielding comparable product titers (Table 1).<br />

Summary: In summary, CAP-T cells present a highly efficient transient<br />

expression system enabling the generation of mg amounts of the protein<br />

of interest for early research and development within only two weeks<br />

from gene to product. Furthermore, CAP-T cell produced proteins showed<br />

fully human posttranslational modification pattern, which was also<br />

observed for the original human CAP cells, the CAP-T cells were derived<br />

from. The CAP technology based on CAP-T cells for transient transfection<br />

and CAP cells for stable protein production [1] therefore provides a<br />

unique system in which the whole process from early research to<br />

production of therapeutic proteins can be run through with the same cell<br />

type.<br />

Reference<br />

1. Essers R, Kewes H, Schiedner S: Improving volumetric productivity of a<br />

stable human CAP cell line by bioprocess optimization. BMC Proceedings ,<br />

abstract within the same supplement.<br />

Table 1(abstract P133) summarizes the relevant data from transient transfections of CAP-T cells in different scales<br />

with plasmids coding for different proteins of interest (hAAT, erythropoietin (EPO), C1-Inhibitor and IgG). Cells were<br />

either transfected by nucleofection (1 x 10 7 cells) or by PEI (1.7 x 10 9 cells)<br />

hAAT EPO C1-Inhibitor IgG<br />

cells transfected 1 x 10 7<br />

1.7 x 10 9<br />

1x10 7<br />

1x10 7<br />

1x10 7<br />

culture volume [ml] 30 1000 60 30 30<br />

culture time [days] 9 6 10 12 6<br />

viability at harvest [%] 85 70 80 87 80<br />

volumetric productivity [mg/L] 170 180 38 35 150


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

P134<br />

Production and purification of TGFb-1 in CHO-Cells<br />

Estabraq Abdulkerim 1* , Sabrina Baganz 1 , Axel Schambach 2 , Cornelia Kasper 1 ,<br />

Thomas Scheper 1<br />

1 Institute of Technical Chemistry, Leibniz University of Hanover, 30167<br />

Hannover, Germany; 2 Department of Experimental Hematology, Hanover<br />

Medical School, 30625 Hannover, Germany<br />

BMC Proceedings 2011, 5(Suppl 8):P134<br />

Introduction: The development of chemically well defined media is a<br />

demanding task in order to create the optimal conditions for an in vitro<br />

stem cell (SC) proliferation and differentiation system. Signals that govern SC<br />

differentiation into multiple mature cell types are provided by growth<br />

factors. TGFb-1 regulates a number of biological processes, including cell<br />

differentiation and proliferation, embryonic development, apoptosis and<br />

immune responses, together with cell surface receptors and signal<br />

transduction molecules within the cell. The whole signal transduction<br />

pathway leads to the specific production of distinct proteins. In this work,<br />

TGFb-1 fragment (A280 – S391) is produced using a tailor-made CHO cell<br />

line (CHO SFS ) and purified.<br />

Results: Chinese Hamster Ovary cells have been subjected to lentiviral<br />

transduction of TGF beta 1 vector (figure 1) resulting in the expression<br />

of His- and HA-tagged protein from (CHO SFS ) cells. This transduction<br />

was accomplished at the department of Hematology, Hanover Medical<br />

School [1].<br />

Production test for TGFb-1: The cells were verified via flow cytometry for<br />

the successful transduction. The selected method involves the specific<br />

Figure 1(abstract P134) TGFb-1 vector used for the lentiviral transduction of CHO cell line (CHO SFS ).<br />

Page 180 of 181<br />

intracellular detection of His-tag by the help of fluorescence-labelled<br />

antibody.<br />

The fixation of 2*10 5 cells was performed by 4 % paraformaldehyde solution,<br />

cells were permeated with 0.1 % saponin followed by an incubation of the<br />

cells in 100 µL primary antibody. Afterwards the antibody was coupled to a<br />

Phycoerythrin (PE) labelled secondry antibody with a fluorescent character.<br />

To control staining specifity, non trasfected CHO cells were used as<br />

negative control which was treated with the same procedure as<br />

transfected cells. The results of the staining show that about 77% of the<br />

cells are successfully transfected.<br />

For the localisation of TGFb-1 cell culture supernatant and cell pellets after<br />

lysis via ultrasonic, were analysed via western blot using a combination of<br />

mouse-anti-His-tag and goat-anti-mouse-IgG-AP-conjugate. The results<br />

indicate that only a part of the protein is secreted extracellulary and the rest<br />

is present intracellulary.<br />

Cultivation of CHO cell line (CHO SFS ): CHO cells were cultured in serum<br />

free ProCHO 5 with 1% penicillin/streptomycin (PAA) and 2% L-glutamine<br />

solution (4mM). A 250 ml spinner flask (rpm. 80) was used for cell growth<br />

starting with a cell density of 0.4 * 10 6 cells*ml -1 and a volume of 100 ml.<br />

The cultivation was carried out for 108 hours and medium was changed<br />

every 2-3 days. Samples were taken every 24 hour to determine cell<br />

density and viability. A maximal cell density of 1.8*10 6 cells/ml could be<br />

achieved with a viability of 80 %.<br />

Purification of TGF beta 1: The supernatant of the culture was used, to<br />

perform the downstream processing.<br />

The cells were separated by means of centrifugation and the clean<br />

supernatant was purified via heparin affinity chromatography (HiTrap TM<br />

Heparin HP columns, GE Healthcare).


BMC Proceedings 2011, Volume 5 Suppl 8<br />

http://www.biomedcentral.com/1753-6561/5?issue=S8<br />

The elution was performed using the binding buffer (10 mM NaH 2PO 4,<br />

pH 7) in addition to a linear NaCl gradient (max. 2M).<br />

Afterwards all protein containing fractions obtained by FPLC were<br />

analysed using silver stained SDS-PAGE. The results show that TGFb-1<br />

could be purified with a purity of 70-80 %.<br />

Conclusion: The production and secretion of TGFb-1 in the CHO cell line<br />

(CHO SFS ) was successfully performed. The purification of protein was<br />

achieved using heparin affinity chromatography. Further upscaling of the<br />

procedure will be performed for achieving higher yield of the targeted<br />

protein.<br />

Acknowledgement: This work was performed within the activities of the<br />

JRG “large scale cultivation” of the DFG cluster of excellence “Rebirth”.<br />

Page 181 of 181<br />

Reference<br />

1. Schambach A, Galla M, Modlich U, Will E, Chandra S, Reeves L, Colbert M,<br />

Williams DA, von Kalle C, Baum C: Lentiviral vectors pseudotyped with<br />

murine ecotropic envelope: Increased biosafety and convenience in<br />

preclinical research. Experimental Hematology 2006, 34:588-592.<br />

Cite abstracts in this supplement using the relevant abstract number,<br />

e.g.: Abdulkerim et al.: Production and purification of TGFb-1 in CHO-Cells.<br />

BMC Proceedings 2011, 5(Suppl 8):P134

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!