bioprocessing of recombinant cho-k1, cho-dg44 and cho...

36
1 Research Article Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO-S: CHO expression hosts favor either mAb production or biomass synthesis David Reinhart 1,* , Lukas Damjanovic 1 , Christian Kaisermayer 2 , Wolfgang Sommeregger 3 , Andreas Gili 4 , Bernhard Gasselhuber 5 , Andreas Castan 6 , Patrick Mayrhofer 1 , Clemens Grünwald-Gruber 5 , Renate Kunert 1 1 Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 11, 1190 Vienna, Austria 2 BioMarin International Limited, Shanbally, Ringaskiddy, County Cork, Ireland 3 Bilfinger Industrietechnik Salzburg GmbH, Urstein Nord 31, 5412 Puch bei Hallein, Austria 4 Polymun Scientific Immunbiologische Forschung GmbH, Donaustraße 99, 3400 Klosterneuburg, Austria 5 Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria 6 GE Healthcare Life Sciences AB, Björkgatan 30, 75184 Uppsala, Sweden *Correspondence: Dr. David Reinhart Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 11, 1190 Vienna, Austria Email: [email protected] This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: [10.1002/biot.201700686]. This article is protected by copyright. All rights reserved Received: November 6, 2017 / Revised: April 9, 2018 / Accepted: April 12, 2018

Upload: others

Post on 18-Nov-2020

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

1

Research Article

Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO-S: CHO expression hosts

favor either mAb production or biomass synthesis†

David Reinhart1,*, Lukas Damjanovic1, Christian Kaisermayer2, Wolfgang Sommeregger3,

Andreas Gili4, Bernhard Gasselhuber5, Andreas Castan6, Patrick Mayrhofer1, Clemens

Grünwald-Gruber5, Renate Kunert1

1Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna,

Muthgasse 11, 1190 Vienna, Austria

2BioMarin International Limited, Shanbally, Ringaskiddy, County Cork, Ireland

3Bilfinger Industrietechnik Salzburg GmbH, Urstein Nord 31, 5412 Puch bei Hallein, Austria

4Polymun Scientific Immunbiologische Forschung GmbH, Donaustraße 99, 3400

Klosterneuburg, Austria

5Department of Chemistry, University of Natural Resources and Life Sciences, Vienna,

Muthgasse 18, 1190 Vienna, Austria

6GE Healthcare Life Sciences AB, Björkgatan 30, 75184 Uppsala, Sweden

*Correspondence: Dr. David Reinhart

Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna,

Muthgasse 11, 1190 Vienna, Austria

Email: [email protected]

†This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: [10.1002/biot.201700686]. This article is protected by copyright. All rights reserved Received: November 6, 2017 / Revised: April 9, 2018 / Accepted: April 12, 2018

Page 2: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

2

Authors

David Reinhart: [email protected]

Lukas Damjanovic: [email protected]

Wolfgang Sommeregger: [email protected]

Andreas Gili: [email protected]

Bernhard Gasselhuber: [email protected]

Andreas Castan: [email protected]

Christian Kaisermayer: [email protected]

Patrick Mayrhofer: [email protected]

Clemens Grünwald-Gruber: [email protected]

Renate Kunert: [email protected]

Keywords: Batch; bioprocess development; Chinese hamster ovary; fed-batch; monoclonal

antibody; perfusion; product quality

Abbreviations: BAC, bacterial artificial chromosome; CHO, Chinese hamster ovary; HC,

heavy chain; LC, light chain; mAb, monoclonal antibody; STY, space-time yield; VCCD,

viable cumulative cell days

Page 3: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

3

Abstract

Chinese hamster ovary (CHO) cells comprise a variety of lineages including CHO-DXB11,

CHO-K1, CHO-DG44 and CHO-S. Despite all CHO cell lines sharing a common ancestor,

extensive mutagenesis and clonal selection has resulted in substantial genetic heterogeneity

among them. Data from sequencing shows that different genes are missing in individual CHO

cell lines and each cell line harbors a unique set of mutations with relevance to the bioprocess.

However, not much literature is available about the influence of genetic differences of CHO

on the performance of bioprocess operations. In this study, we examined host cell-specific

differences among three widely used CHO cell lines (CHO-K1, CHO-S and CHO-DG44) and

recombinantly expressed the same monoclonal antibody (mAb) in an isogenic format by using

bacterial artificial chromosomes (BACs) as transfer vector in all cell lines. Cell-specific

growth and product formation were studied in batch, fed-batch and semi-continuous perfusion

cultures. Further, two different cell culture media were used to investigate their effects. We

found CHO cell line-specific preferences for mAb production or biomass synthesis that were

determined by the host cell line. Additionally, quality attributes of the expressed mAb were

influenced by the host cell line and media.

Page 4: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

4

1. Introduction

CHO cells comprise a variety of lineages such as CHO-DXB11 (or DUKX), CHO-K1, CHO-

DG44 and CHO-S that share a common ancestor. The original CHO cell line was generated

by Dr. Theodore Puck in 1956 who isolated spontaneously immortalized fibroblasts from a

culture of ovarian cells of a Chinese hamster [1]. CHO-K1 was derived from a subclone of the

original cell line in 1957. In 1980, CHO-DXB11 (or DUKX) was generated through chemical

mutagenesis of CHO-K1 [2]. The dihydrofolate reductase (dhfr) deficiency enabled the stable

introduction of transgenes when co-transfected with a functional copy of the dhfr gene as a

selection marker. In 1983, Urlaub et al. [3] deleted both dhfr alleles by mutagenesis of a

different CHO cell starting population, thus generating CHO-DG44 lineage. In 1991, the

CHO-S cell line was generated from another CHO cell starting population. Nowadays, all

these cell lines are widely used for the production of biopharmaceuticals.

Since the introduction of the original CHO cell line, extensive mutagenesis and clonal

selection has resulted in substantial genetic heterogeneity among the different CHO cell

lineages [4-6]. Karyotyping of various CHO cells underpins this genetic diversity by

highlighting several deletions, translocations or rearrangements of chromosomal segments

[4]. Even within a supposedly monoclonal population of the ancestor of CHO-K1, substantial

heterogeneity was found [7]. Similar observations were made in a karyogram of CHO-DG44

cells [8]. Further evidence concerning phenotypic diversity comes from substantial gene copy

number variations (CNVs), ploidy and small-nucleotide polymorphisms (SNPs) among CHO-

K1, CHO-DG44 and CHO-S lineages [5]. Finally, epigenetic variations (e.g. histone

modifications, chromatin remodeling and DNA methylation) modifying gene expression also

exists for CHO cells [9-11].

In recent years, sequencing data demonstrated genetic differences among different CHO cell

lines leading to speculation on their relevance for bioprocessing [4, 5, 12-14]. However, while

sequencing data increases, studies investigating their biological/technological significance

Page 5: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

5

remain scarce. In this study, we examined host cell-specific differences of production clones

among three widely used CHO cell lines (CHO-K1, CHO-S and CHO-DG44). We expressed

the same mAb in all three CHO cell lines to avoid product-related effects on the host cell line

such as the physicochemical properties of the recombinant product [15] or on nutrient

requirements (e.g. amino acid usage). Position effects, which are commonly observed upon

random integration of plasmid DNA, were circumvented by using BACs as genetic transfer

vehicle providing a quasi-isogenic environment of the recombinant gene in different

production clones. These vectors achieve position-independent, but gene copy number

(GCN)-dependent, high and stable product expression even without the need of transgene

amplification [16]. Cell growth and product formation were studied in batch, fed-batch and

semi-continuous perfusion cultures. Two different cell culture media were used for all cell

lines to investigate the effect on the bioprocess and mAb quality. Phenotypic differences

among CHO cell lineages were confirmed in our study demonstrating the effect of genotypic

variations and their relevance for bioprocessing. The combination of CHO sequence-specific

knowledge and lineage-specific cultivation behavior will be a powerful tool for future cell

line engineering and development, as well as improvement of bioprocesses and cell culture

media.

Page 6: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

6

2. Materials and methods

2.1. Cell lines and media

2.1.1. Generation of recombinant cell lines

Recombinant mAb (IgG) expressing CHO-K1, CHO-DG44 and CHO-S cell lines were

generated according to a previously described methodology [16]. Briefly, serum-free adapted

host cell lines derived from CHO-K1 (ATCC CCL-61), CHO-DG44 (Life Technologies) and

CHO-S (Life Technologies) were transfected using linearized BAC DNA, containing

antibody light and heavy chain transgenes, and Lipofectin (Life Technologies). In-between

the first thaw of the cells upon acquisition and their transfection, host cells were passaged

under controlled conditions 10 (CHO-S), 62 (CHO-K1) and 89 times (CHO-DG44). The

primary transfectants were seeded as mini-pools (5,000 cells/well in 100 µL) in 96-well

culture plates (Nunc, Thermo Fisher Scientific) and incubated at 37°C and 5% CO2 for 2-3

weeks. Then, wells in which cell growth was observed were screened for mAb production.

Mini-pools with high mAb concentrations were expanded in T-25 cell culture flasks (Greiner

Bio-One). During three consecutive passages, the best clone was selected based on cell-

specific mAb productivity (qP) and cell growth for single-cell dilution sub-cloning (one

cell/well in a 384-well cell culture plate). Grown (monoclonal) wells were expanded and

analyzed as described above to define the final production clone. CHO-DG44 host cell lines

are deficient in the dhfr gene, which is typically co-transfected with the gene of interest

during cell line development. Since CHO-K1 and CHO-S cells produce dhfr, we co-

transfected the CHO-DG44 cells with the BAC and a dhfr-containing plasmid for a fair

evaluation.

2.1.2. Medium adaptation and cell banking

All CHO cell lines were initially grown in CD CHO (Life Technologies) supplemented with 8

mM L-glutamine (Sigma-Aldrich) and 0.5 mg/mL G418 (Sigma-Aldrich) during routine

Page 7: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

7

cultivation. Routine cultures were inoculated at a cell concentration of 3×105 c/mL in 125 mL

Erlenmeyer shake flasks (Corning) at a working volume of 35 mL and cultivated in an ISF1-

X incubator shaker (Kuhner) operated at 37°C, 140 rpm, 7% CO2 and 90% relative humidity.

The cells were split every 3 to 4 days.

Adaptation to ActiCHOTM P (GE Healthcare) was performed by direct inoculation into the

alternative medium. When the cells reached similar concentrations and viabilities as in CD

CHO, the homogeneity (absence of subpopulations) of the successfully adapted cell lines was

confirmed by flow cytometry according to a previously published protocol [17]. The absence

of any low or non-producing subpopulations was confirmed by visualization of intracellularly

stained antibody heavy and light chains with FITC-conjugated anti-human γ-chain (Sigma)

and Alexa Fluor-647-conjugated anti-human λ-chain specific antiserum (Thermo Scientific),

respectively. Subsequently, research cell banks were prepared in CryoMaxx II (GE

Healthcare) for all following experiments.

2.2. Bioprocessing in batch, fed-batch and perfusion mode

Batch and fed-batch experiments were performed in a shake incubator at the conditions

described above. 500 mL Erlenmeyer shake flasks (Corning) were inoculated at a starting cell

concentration of 3×105 c/mL and a working volume of 130 mL using ActiCHO P or CD CHO

as basal media. Cultures were terminated once the viability dropped below 60%. All

experiments were conducted as biological triplicates from the same inoculum and operated as

individual cultivations.

Batch experiments were analyzed daily for cell concentration, viability, antibody and

metabolite concentrations, as well as osmolality.

In fed-batch experiments, two strategies were compared using different basal and feed media.

In ActiCHO P, nutrient feeding started on day 3 with daily feed additions of ActiCHOTM Feed

A and ActiCHOTM Feed B (both GE Healthcare) at 3% and 0.3% of the working volume,

Page 8: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

8

respectively. In CD CHO, cultures were fed on days 4, 6 and 8 with CHO CD

EfficientFeedTM A and FunctionMAXTM Titer Enhancer (both Life Technologies) at 10% and

3.3% of the working volume, respectively. For both fed-batch strategies, the residual glucose

concentration was maintained above 3 g/L by adding a 250 g/L concentrated glucose solution

(Sigma-Aldrich) to a final concentration of 6 g/L. Sampling was performed as described for

batch experiments. Total RNA was prepared at day 4, 7 and at harvest to analyze intracellular

heavy chain and light chain mRNA levels by qPCR. Product quality was analyzed by mass

spectrometry (glycosylation) and differential scanning calorimetry (thermal stability).

Semi-continuous perfusion experiments were conducted at conditions described above at a

starting cell concentration of 5×106 c/mL. The basal cell culture medium was exchanged once

per day after separating the cells by centrifugation applying 200×g for 10 min. The

centrifugation conditions are commonly applied for routine cultivation and do not impact cell

growth or viability. The semi-continuous perfusion cultures were operated for 11 days at high

viabilities and then terminated.

2.3. Cell concentration and viability

The total cell concentration was quantified using a Z2 Coulter Counter™ (Beckman Coulter)

according to the manufacturer’s instructions. The viability was determined by trypan blue dye

exclusion with a Neubauer improved hemocytometer (MedPro).

2.4. Antibody concentration

Antibody concentrations were determined by Bio-Layer Interferometry on an Octet™ QK

(Pall) as previously described [18].

Page 9: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

9

2.5. qPCR

Preparation of cDNA was previously described [19]. qPCR was performed using the

MiniOpticon™ system with 48-well low white PCR plates and microseal B film sealer (all

from BioRad). All primers were designed using the Primer3 web application [20] and

synthesized (Sigma-Aldrich). Primers were designed to amplify fragments of the nt-sequence

in the constant regions of the mAbs heavy and light chain as shown in Tab.1. The genes of β-

actin (ACTB) and the eukaryotic translation initiation factor 3 subunit I (EIF3I) served as

internal reference genes. qPCR was performed with 3 ng of cDNA including non-template

controls, negative controls and no-reverse-transcriptase controls. Next to the template, each

reaction mix contained 10 µL KAPA PROBE Fast Universal (peqlab) and 6 pmol of each

primer. qPCR was performed in 2-step mode and all samples were measured in triplicates of

three biological samples in two technical runs. Data evaluation and calculations based on the

2−ΔΔCq method [21] were previously described [19, 22]. Data normalization was performed

using both reference genes.

2.6. Antibody purification

For analysis of antibody product quality, cell culture supernatants were purified using

HiTrap™ MabSelect SuRe™ 1 mL columns according to the manufacturer’s

recommendations (GE Healthcare).

2.7. Glycan analysis

Glycoprofiling was performed to investigate oligosaccharide distribution of antibody product

upon harvest from fed-batch cultures. The cell suspension was centrifuged at 170×g, the

supernatant clarified using a 0.22 µm filter (Express PLUS, Merck Millipore) and diluted with

PBS to a concentration of 1 to 2 mg/mL. N-glycan profiles of the Fc regions of the

monoclonal antibodies were determined by IdeS proteolytic digestion and electrospray mass

Page 10: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

10

spectrometry according to a published protocol [23]. This procedure was modified by

including carboxypeptidase B to remove unprocessed C-terminal lysine residues to improve

the quality of the data obtained for the Fc region of the heavy chain. Also, a reduction step

was introduced to improve the quality of data obtained for the light chain and the heavy chain

Fab region. Analysis was performed using the separation offered by reversed phase liquid

chromatography in combination with detection by electrospray Q-Tof mass spectrometry. Fc

fragment signals having masses corresponding to N glycan isoforms G0, G0F, G0F-Gn, G1F-

Gn, G1F, G2F, G1F+SA1, G2F+SA1, G2F+SA2, Man5 to Man9, Man9+Glc, and Man-

9+2Glc were investigated and their relative abundance estimated from the intensity of these

signals. The small signals for N glycan isoforms G1 and G2 were not amenable to

quantification as their mass is only separated by 6 Da from the sodium adducts of G0F and

G1F isoforms.

2.8. Differential Scanning Calorimetry

Thermal denaturation of antibody samples was analyzed using automated differential

scanning calorimetry (DSC). All DSC measurements were performed on a VP-DSC MicroCal

LLC equipment (GE Healthcare). Protein solutions were sampled from 96-well plates using

the robotic attachment. Protein concentration of all samples was 10 μM. The temperature

profile was recorded between 20°C and 100°C with a scan rate of 1°C/min. The results were

evaluated and fitted with Origin 7.0 software (OriginLab). The unfolding states of the

antibodies were fit using the non-two state unfolding model in the software.

3. Results

3.1. Generation of recombinant CHO cell lines and media adaptation

Unlike randomly integrated plasmid DNA, which often results in transfectants with

substantially differing rates of recombinant protein expression, BAC based vectors enable a

Page 11: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

11

more comparable and stable product expression even without the need of transgene

amplification. For this study, three recombinant CHO cell lines were generated by

transfection with the same BAC constructs in CHO-K1, CHO-S and CHO-DG44 host cell

lines. Among all screened mini-pools, the average specific mAb production rates prior to

subcloning were 5.04 ± 2.37 (CHO-K1), 3.29 ± 1.82 (CHO-S) and 1.10 ± 0.73 (CHO-DG44)

picogram mAb per cell and day (pcd).

Cell line development and clone selection was initially performed in CD CHO. To investigate

potential medium-specific effects, all three CHO cell lines were adapted to a second

chemically defined cell culture medium (ActiCHO P). Direct adaptation was successful and

all three recombinant CHO cell lines reached similar or even higher cell concentrations and

viabilities during routine cultivation as in the initial medium (Supplement 1). Furthermore, a

successful medium adaptation was confirmed by affirming the homogeneity of all adapted

cell lines and the absence of any low or non-producing subpopulations (Supplement 2).

3.2. Batch evaluation of CHO-K1, CHO-S and CHO-DG44 cell lines

Batch cultivation revealed higher peak cell concentrations for CHO-S and CHO-DG44 than

for CHO-K1 (Fig.1; Supplement 3). CHO-S cells grew with the highest cell-specific growth

rate (Tab.2), reached the maximum cell concentration first and had the shortest process

duration (Fig.1). Higher cell concentrations were obtained using ActiCHO P. CHO-K1

showed 4 to 6-fold increased cell-specific mAb production rates compared with CHO-S and

CHO-DG44 during batch culture. Consequently, these cell cultures reached the highest

product titers and CHO-S the lowest titers in both media. The diagram plotting the VCCD

against mAb titer (Fig. 1) demonstrated CHO cell-specific preferences for fast growth (CHO-

S) or recombinant protein production (CHO-K1). In batch culture, no major difference in titer

was observed for the two cell culture media tested. The difference between the cell lines was

more pronounced than the difference resulting from cultivation media.

Page 12: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

12

3.3. Fed-batch evaluation of CHO-K1, CHO-S and CHO-DG44 cell lines

In fed-batch cultivation, CHO-S cells also grew with the highest cell-specific growth rates

during the initial five days (Tab.2; Supplement 3). The final mAb titer was generally higher in

ActiCHO P with CHO-DG44 reaching 670 mg/L followed by CHO-K1 (460 mg/L) and

CHO-S (370 mg/L). In CD CHO fed-batches, CHO-K1 cells (350 mg/L) produced higher

titers than CHO-S (175 mg/L) and interestingly the lowest mAb concentration was

determined in cultures of CHO-DG44 (115 mg/L) because of poor biomass accumulation and

low mAb production rates (Fig.1, Tab.2). These results demonstrated that the addition of feed

concentrates substantially influenced cell accumulation by favoring one CHO host over the

other regarding their specific nutrient demands. Nevertheless, cell-specific mAb production

rates remained highest in CHO-K1 followed by CHO-DG44 and CHO-S (Tab.2.). This was

also observable from Fig.1 showing CHO cell-specific preferences for either biomass

synthesis (CHO-S) or recombinant protein production (CHO-K1).

Compared to batch cultivation, feed supplementation of CHO-K1 cells in ActiCHO P resulted

in doubling peak cell concentrations but only 10-20% higher titers due to a significant

reduction of cell-specific mAb production rates. Feed supplementation of CHO-S cells did not

significantly impact peak cell concentrations. However, due to a substantially prolonged

process duration by six days, VCCD increased significantly and boosted mAb titers by more

than six-fold. In CD CHO, feed supplementation of CHO-S cells was less effective in

increasing VCCD and process duration than in ActiCHO P. Thus, final titers improved only

2.5-fold compared to batch cultivation. Fed-batch cultivation of CHO-DG44 cells doubled

peak cell concentrations, VCCD and cell-specific production rates compared to batch cultures.

This improved mAb titer more than five-fold. Interestingly, feed supplementation in CD CHO

did not improve the process performance compared to batch cultivation.

Page 13: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

13

3.4. Perfusion evaluation of CHO-K1, CHO-S and CHO-DG44 cell lines

To study the CHO cell lines under quasi steady-state conditions we used a lab-scale semi-

continuous perfusion system with daily media exchange (Fig.1; Supplement 3). CHO-S cells

grew with the highest cell-specific growth rate (Tab.2) and cell concentration in theses

cultures peaked first (Fig.1). CHO-K1 cells plateaued among the lowest peak cell

concentrations (~25×106 c/mL) at values comparable to CHO-DG44 cells cultivated in CD

CHO. Interestingly, CHO-DG44 cell concentrations continuously increased to the highest

values of all CHO cells when cultivated in ActiCHO P. These results demonstrated that daily

media exchange considerably affects growth of CHO host cell lines due to their individual

nutrient demands. Upon media exchange every 24 hours, the highest mAb concentrations

were generated by CHO-K1 cells followed by CHO-DG44 cells and CHO-S cells. In both

media, CHO-K1 cells had the highest cell-specific mAb production rates (13-16 pcd), thus

outcompeting CHO-DG44 cells (4-5 pcd) and CHO-S cells (ca. 2 pcd). These results aligned

well with the qP values for the different cell lines in batch and fed-batch cultures (Tab.1).

3.5. Evaluation of mRNA

To investigate if high cell-specific mAb production rates coincided with high heavy and light

chain mRNA levels, we analyzed both parameters on several days during fed-batch

cultivation. For all three CHO cell lines, cell-specific mAb production rates declined

continuously, while HC and LC mRNA accumulated with increasing variability between

biological triplicates (Figure 2). Therefore, we statistically evaluated product-specific mRNA

and qP on day 4 when all clones had comparable cell densities and cultures were still in the

exponential growth phase. Interestingly, HC and LC mRNA in CHO-K1 cells were

significantly influenced by the feed medium and this was reflected in the specific productivity

(Supplement 4). Statistical evaluation gave also evidence that HC mRNA contributed more

directly to a higher qP than LC mRNA.

Page 14: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

14

3.6. CHO host cell line and mAb product quality

The observed phenotypic differences during cultivation of the different CHO cell lines

prompted us to analyze the secreted product. We decided to investigate mAb product from

fed-batch cultivation harvest only, first, because most currently executed bioprocesses rely on

this strategy and second, we speculated that due to product accumulation in fed-batch even

minor cell line-specific product-related differences might become more visible compared to

other process modes.

In general, all three CHO cells expressed mainly core fucosylated complex biantennary Fc

glycans with predominantly G0F, G1F and G2F glycoforms (Fig.3), similar to naturally

occurring human serum IgG [24]. The relative abundance of some glycoforms was influenced

by CHO cell line or media. For example, when the media was changed from ActiCHO P to

CD CHO, the amount of galactosylated mAb increased from 17% to 37% for CHO-K1 and

CHO-S, but remained constant for CHO-DG44. The abundance of agalactosylated glycoforms

(G0F) was higher for CHO-S than for CHO-K1 and CHO-DG44 expressed mAb. Product

mannosylation, fucosylation and aglycosylation were less influenced by the media, but merely

an inherent trait of the CHO cell line. CHO-S cells expressed the lowest amount of

mannosylated product (2-3%) relative to CHO-K1 (5-9%) and CHO-DG44 cultures (11-

13%). Product fucosylation was highest in CHO-S (94-96%) followed by CHO-K1 (82-84%)

and CHO-DG44 (71-83%). The amount of aglycosylated mAb was low in CHO-K1 (2-5%)

and CHO-S (1%) and no aglycosylated mAb was detected in CHO-DG44 cultures.

In addition to the Fc glycosylation site, the herein expressed mAb contained a further glycan

moiety in the light chain region. This glycosylation site also contained mainly core

fucosylated complex biantennary glycans with predominantly G0F, G1F and G2F glycoforms,

but occasionally one or two sialic acid residues (Fig.4). CHO-S cells expressed the lowest

amount of mannosylated product (≤ 1.5%) relative to CHO-K1 (4-6%) and CHO-DG44

Page 15: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

15

cultures (5-13%). Product fucosylation of the light chain was lowest in CHO-S (~46%)

followed by CHO-DG44 (76-88%) and CHO-K1 (83-90%). The amount of sialylated mAb

was highest in CHO-K1 (21-36%) followed by CHO-DG44 (15-19%) and CHO-S cultures (6-

15%). Despite detectable changes of mAb glycosylation they were subtle and their

pharmacological relevance would need to be evaluated in more detail.

Mammalian cells commonly perform post-translational modifications during protein

production that are essential for protein folding, stability and function [25, 26]. To investigate

conformational stabilities of the same mAb produced by different CHO hosts we analyzed

thermal stabilities of the protein. DSC measurements revealed four unfolding transitions for

the investigated mAbs (Fig.5). Most important, the media used for cultivation did not

influence the thermal stability of expressed mAb. Remarkably, the first mAb unfolding

transition (Tm1) started 1.7 °C earlier when expressed by CHO-S (64.3 °C ± 0.2) compared to

mAb expressed in CHO-K1 or CHO-DG44 (both 66.0 °C ± 0.3). To further investigate

potential reasons for the differences of thermal stability we analyzed the amino acid sequence

of mAb product by mass spectrometry (data not shown) and found that the mAb expressed in

CHO-S (but not CHO-K1 and CHO-DG44) missed the C-terminal serine in the light chain.

4. Discussion

To date, more than 70% of all recombinant biopharmaceuticals are expressed in CHO cells

[27]. However, current literature is scarce in CHO host comparisons, suggesting that such

knowledge can be an advantage. It is not surprising that companies rely on individually

selected host cell line(s) to ensure repeatedly reaching the desired product quantity and

quality attributes of any to-be-expressed biomolecule. To shed more light on this topic, we

investigated growth and recombinant mAb expression of three commonly used CHO cell lines

in different cultivation modes. The herein presented study demonstrated host-specific

physiologic preferences favoring either biomass synthesis or antibody production (Fig.1).

Page 16: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

16

4.1. CHO host favors mAb production or biomass synthesis

For a proper comparison of cell lines that were indeed representative for the respective CHO

host, we have applied the same transfection and selection criteria for all three monoclonal cell

lines. Only the best producers with adequate growth properties were selected according to

good practice in cell line generation.

Already during cell line generation and screening of polyclonal mini-pools, CHO-K1 cells

revealed a higher mean qP than CHO-S and CHO-DG44. This observation was persistent

from the initial mini-pools until the finally derived monoclonal cell lines. Combined with the

consistent genetic environment provided by the BAC constructs, we firmly believe that the

reported results are due to phenotypic differences rather than clonal variations.

Cell culture media are complex nutrient solutions that contain up to 100 different components

at varying concentrations. This makes medium adaptation complex and may affect cell

growth, viability, protein expression and even genomic stability [18, 28, 29]. In our case,

direct adaptation was successful for all three CHO cell lines without introducing any lag

phases or lowering the viability (Supplement 1). Despite genetic changes were not

investigated, the absence of low or non-producing subpopulations was confirmed upon

medium adaptation (Supplement 2).

Cell growth and peak cell concentrations were generally higher in ActiCHO P (Tab.2) during

all process strategies (i.e. batch, fed-batch and semi-continuous perfusion cultures) indicating

that a balanced media formulation can influence the performance of the cultivated cell lines

[18]. Independent of medium or process type, CHO-S cells grew with 10-50% higher cell-

specific growth rates than CHO-K1 and CHO-DG44 cells (Tab.2). This highlighted CHO

host-inherent traits towards individual preferences for cell growth. Interestingly, also the non-

transfected CHO-S host cell line reached higher growth rates than CHO-K1 and CHO-DG44

during batch and routine cultivation.

Page 17: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

17

CHO-K1 cells had the highest cell-specific productivities as indicated by high mAb titers with

relatively low biomass accumulation. CHO-S showed opposite characteristics (Tab.2, Fig.1).

CHO-DG44 ranked in-between the two other CHO cell lines. Remarkably, this trend was

consistent in all cultivation modes and cell culture media. Determination of GCN showed

between 1-2 LC copies and 2-4 HC copies in the different CHO cell lines despite both cDNAs

were part of the same BAC. This was most likely a result of the underlying logarithmic

calculation. In our dataset, we found no statistically significant correlation of product-related

mRNA to cell-specific mAb production rates over the whole process since both parameters

changed with process time for all three CHO cell lines (Figure 2). Considering the extensive

mutagenesis and clonal selection of the derived CHO host cell lines [4], we assume that the

determined expression differences related to the cellular phenotype and metabolism rather

than differences in product-specific GCN (or mRNA), location of the transgenes [12], or type

of cell cultivation. The influence of GCN and amount of transcript on protein expression

levels is addressed since a long time by several publications and remains controversial [30-

34].

Differing rates of product synthesis among the CHO host cell lines could be due to unequal

capacities and processing efficiencies of their translational machineries. For example, a larger

ER and higher mitochondrial mass of CHO-K1 has recently been associated with a higher qP

compared to CHO-DUXB11 [35]. The ER is important for proper folding and assembly of

secreted proteins. If the amount of irreversibly misfolded proteins in the ER exceeds a certain

limit, cells will trigger the unfolded protein response and eventually apoptosis [36, 37]. A

larger ER thus favors higher secretion rates but also the survival of high-producing cells and

the processing of difficult-to-fold proteins. Mitochondria provide the energy to many cellular

processes including protein folding [38]. A higher mitochondria mass may be advantageous to

cells in order to self-fuel their required energy to cope with the resulting stress of misfolded

proteins in the ER. Since both ER and mitochondria occupy a large proportion of the total

Page 18: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

18

cellular volume, capacities for recombinant protein expression may be reflected by cell size.

Interestingly, we observed a larger cell size for high producing CHO-K1 cells (15.7 µm ±0.7)

compared to low producing CHO-DG44 (14.2 µm ±0.5) and CHO-S cells (13.7 µm ±1.1)

during semi-continuous perfusion cultivation.

4.2. CHO host influences mAb glycosylation profile

The glycosylation profile considerably determines mAb safety and efficacy [39]. Changes in

Fc glycosylation can alter Fc conformation, receptor binding and immune effector functions

as reviewed in detail elsewhere [40].

Our study showed that mAb glycosylation was influenced by the applied CHO expression

host (Fig.3 and 4). Mannosylation was higher and fucosylation was lower when mAb was

expressed by CHO-DG44 followed by CHO-K1 and CHO-S. Antibodies with high-mannose

and low-fucose content generally increase ADCC [41, 42]. Galactosylation, which increases

IgG effector functions [43], was merely influenced by the applied cultivation media. Media

constituents that affect mAb galactosylation include uridine, manganese chloride and

galactose [44, 45]. However, their concentrations in the applied cell culture media were not

quantified. In general, CHO-K1 revealed a higher degree of mAb galactosylation than CHO-

DG44 followed by CHO-S. Only carbohydrates of the LC (but not Fc part) contained sialic

acid residues indicating that sialylation was more a consequence of steric accessibility.

Sialylation is often associated with improved serum half-life [46] or anti-inflammatory

activity [47]. Sialylation of mAb was higher for CHO-K1, followed by CHO-DG44 and then

CHO-S cells. Host-specific glycosylation patterns may be related to the heterogeneity of CHO

cells that harbor and express a different set of genes relevant for glyco-processing [5, 13].

Lewis et al. [5] have shown that among 256 enzymes associated with glycosylation in

Cricetulus griseus, 13%, 2% and 25% have non-synonymous SNPs, frame-shifting indels, or

CNVs, respectively, in at least one cell line of CHO-K1, CHO-DG44 and CHO-S. While not

Page 19: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

19

all SNPs will have a measurable effect on enzyme activity or substrate preference, glycans are

produced through long, branching pathways, which increases the probability of having a

mutation that changes the final glycoform in any given cell line. This hypothesis supports our

results that different CHO expression systems harbor cell type-specific glycosylation patterns,

which can be exploited depending on the produced protein and its requirements in vivo.

Mammalian cells commonly perform post-translational modifications during protein

production that are essential for protein folding, stability and function [25, 26]. Higher

thermal stabilities improve mAb expression rates [48, 49]. In our study, the least stable mAb

(CHO-S) was expressed at the lowest cell-specific rate (Tab.2). However, the thermal stability

of CHO-DG44 expressed mAb was similar to CHO-K1 despite mAb expression rates were

slightly better than CHO-S. It remains elusive if or to which extent the thermal stability

difference of 1.7 °C (Tm1) contributed to protein expression. A potential reason for lower

stability could be a missing C-terminal serine in the LC of mAb expressed in CHO-S (but not

CHO-K1 and CHO-DG44) as identified by mass spectrometry. If this was due to mAb

processing differences by the CHO host currently remains undetermined. Shen et al. [50]

showed that deletion of the C-terminal serine of the λ LC improved thermal and high pH

stabilities, transient expression, purification yield and ADCC function of an IgG1 without

affecting antigen binding. The herein investigated mAb also was an IgG1 λ, but the thermal

stability was higher when the C-terminal serine was present (Fig.5). The cultivation media did

not influence mAb stability.

5. Conclusion

In recent years, high-throughput technologies have created an ever-increasing amount of

sequencing data that demonstrates the genetic heterogeneity of CHO cell lines. However, the

influence of these genetic differences on the phenotype of different CHO cell lines and finally

the effect on bioprocessing has not yet been investigated in detail. In this study, we found that

Page 20: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

20

the cellular phenotype determines CHO cell line-specific preferences for mAb production or

biomass synthesis. CHO-K1 metabolism favored mAb expression, whereas CHO-S

metabolism had a preference for biomass formation and CHO-DG44 ranked in-between the

two. These preferences were independent of process type (i.e. batch, fed-batch, or perfusion)

and cell culture media. Therefore, the choice of the CHO host cell line had a fundamental

impact on the production process. Currently, genomic and phenotypic variations of CHO cells

are not well understood and often not taken into consideration when selecting a host cell line.

Genetic differences between host cell lines should be examined in biological experiments to

better understand their impact on the cellular phenotype and bioprocess. The combination of

CHO sequence-specific knowledge and lineage-specific cultivation behavior will be a

powerful tool for future cell line engineering approaches and cell line development, as well as

improvement of bioprocesses and cell culture media. The results of this study demonstrate

that the genetic background substantially influences the phenotypic properties of the host cell

also affecting productivity and glycosylation. Therefore, the choice of the host cell line is of

key importance to achieve the desired process characteristics and product quality.

6. Disclosure statement

The authors declare no commercial or financial conflict of interest.

7. Acknowledgements

This study was funded by GE Healthcare. We thank Polymun Scientific Immunbiologische

Forschung GmbH for providing the recombinant cell lines.

Page 21: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

21

8. References

[1] PUCK, T. T., Cieciura, S. J., Robinson, A., Genetics of somatic mammalian cells. III.

Long-term cultivation of euploid cells from human and animal subjects. J Exp Med 1958,

108, 945-956.

[2] Urlaub, G., Chasin, L. A., Isolation of Chinese hamster cell mutants deficient in

dihydrofolate reductase activity. Proc Natl Acad Sci U S A 1980, 77, 4216-4220.

[3] Urlaub, G., Käs, E., Carothers, A. M., Chasin, L. A., Deletion of the diploid dihydrofolate

reductase locus from cultured mammalian cells. Cell 1983, 33, 405-412.

[4] Wurm, F. M., Hacker, D., First CHO genome. Nat Biotechnol 2011, 29, 718-720.

[5] Lewis, N. E., Liu, X., Li, Y., Nagarajan, H., Yerganian, G., O'Brien, E., Bordbar, A.,

Roth, A. M., Rosenbloom, J., Bian, C., Xie, M., Chen, W., Li, N., Baycin-Hizal, D., Latif, H.,

Forster, J., Betenbaugh, M. J., Famili, I., Xu, X., Wang, J., Palsson, B.O., Genomic

landscapes of Chinese hamster ovary cell lines as revealed by the Cricetulus griseus draft

genome. Nat Biotechnol 2013, 31, 759-765.

[6] Xu, X., Nagarajan, H., Lewis, N. E., Pan, S., Cai, Z., Liu, X., Chen, W., Xie, M., Wang,

W., Hammond, S., Andersen, M. R., Neff, N., Passarelli, B., Koh, W., Fan, H. C., Wang, J.,

Gui, Y., Lee, K. H., Betenbaugh, M.J., Quake, S.R., Famili, I., Palsson, B.O., Wang, J., The

genomic sequence of the Chinese hamster ovary (CHO)-K1 cell line. Nat Biotechnol 2011,

29, 735-741.

[7] Deaven, L. L., Petersen, D. F., The chromosomes of CHO, an aneuploid Chinese hamster

cell line: G-band, C-band, and autoradiographic analyses. Chromosoma 1973, 41, 129-144.

[8] Derouazi, M., Martinet, D., Besuchet Schmutz, N., Flaction, R., Wicht, M., Bertschinger,

M., Hacker, D. L., Beckmann, J. S., Wurm, F. M., Genetic characterization of CHO

production host DG44 and derivative recombinant cell lines. Biochem Biophys Res Commun

2006, 340, 1069-1077.

[9] Bauer, A. P., Leikam, D., Krinner, S., Notka, F., Ludwig, C., Längst, G., Wagner, R., The

impact of intragenic CpG content on gene expression. Nucleic Acids Res 2010, 38, 3891-

3908.

Page 22: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

22

[10] Kim, M., O'Callaghan, P. M., Droms, K. A., James, D. C., A Mechanistic Understanding

of Production Instability in CHO Cell Lines Expressing Recombinant Monoclonal

Antibodies. Biotechnol Bioeng 2011, 108, 2434-2446.

[11] Yang, Y. S., Mariati, Chusainow, J., Yap, M. G. S., DNA methylation contributes to loss

in productivity of monoclonal antibody-producing CHO cell lines. J Biotechnol 2010, 147,

180-185.

[12] Kennard, M. L., Goosney, D. L., Monteith, D., Roe, S., Fischer, D., Mott, J., Auditioning

of CHO host cell lines using the artificial chromosome expression (ACE) technology.

Biotechnol Bioeng 2009, 104, 526-539.

[13] Könitzer, J. D., Müller, M. M., Leparc, G., Pauers, M., Bechmann, J., Schulz, P., Schaub,

J., Enenkel, B., Hildebrandt, T., Hampel, M., Tolstrup, A. B., A global RNA-seq-driven

analysis of CHO host and production cell lines reveals distinct differential expression patterns

of genes contributing to recombinant antibody glycosylation. Biotechnol J 2015, 10, 1412-

1423.

[14] Wuest, D. M., Harcum, S. W., Lee, K. H., Genomics in mammalian cell culture

bioprocessing. Biotechnol Adv 2012, 30, 629-638.

[15] Sommeregger, W., Mayrhofer, P., Steinfellner, W., Reinhart, D., Henry, M., Clynes, M.,

Meleady, P., Kunert, R., Proteomic differences in recombinant CHO cells producing two

similar antibody fragments. Biotechnol Bioeng 2016, 113, 1902-1912.

[16] Zboray, K., Sommeregger, W., Bogner, E., Gili, A., Sterovsky, T., Fauland, K., Grabner,

B., Stiedl, P., Moll, H. P., Bauer, A., Kunert, R., Casanova, E., Heterologous protein

production using euchromatin-containing expression vectors in mammalian cells. Nucleic

Acids Res 2015, 43, e102.

[17] Reinhart, D., Sommeregger, W., Debreczeny, M., Gludovacz, E., Kunert, R., In search of

expression bottlenecks in recombinant CHO cell lines-a case study. Appl Microbiol Biot 2014,

98, 5959-5965.

[18] Reinhart, D., Damjanovic, L., Kaisermayer, C., Kunert, R., Benchmarking of

commercially available CHO cell culture media for antibody production. Appl Microbiol

Biotechnol 2015, 99, 4645-4657.

Page 23: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

23

[19] Mader, A., Prewein, B., Zboray, K., Casanova, E., Kunert, R., Exploration of BAC

versus plasmid expression vectors in recombinant CHO cells. Appl Microbiol Biotechnol

2013, 97, 4049-4054.

[20] Untergasser, A., Nijveen, H., Rao, X., Bisseling, T., Geurts, R., Leunissen, J.A.,

Primer3Plus, an enhanced web interface to Primer3. Nucleic Acids Res 2007, 35, W71-74.

[21] Livak, K. J., Schmittgen, T. D., Analysis of relative gene expression data using real-time

quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001, 25, 402-408.

[22] Sommeregger, W., Prewein, B., Reinhart, D., Mader, A., Kunert, R., Transgene copy

number comparison in recombinant mammalian cell lines: critical reflection of quantitative

real-time PCR evaluation. Cytotechnology 2013, 65, 811-818.

[23] Chevreux, G., Tilly, N., Bihoreau, N., Fast analysis of recombinant monoclonal

antibodies using IdeS proteolytic digestion and electrospray mass spectrometry. Anal

Biochem 2011, 415, 212-214.

[24] Jefferis, R., Glycosylation of recombinant antibody therapeutics. Biotechnol Prog 2005,

21, 11-16.

[25] Walsh, G., Jefferis, R., Post-translational modifications in the context of therapeutic

proteins. Nat Biotechnol 2006, 24, 1241-1252.

[26] Zheng, K., Yarmarkovich, M., Bantog, C., Bayer, R., Patapoff, T. W., Influence of

glycosylation pattern on the molecular properties of monoclonal antibodies. MAbs 2014, 6,

649-658.

[27] Jayapal, K. R., Wlaschin, K. F., Hu, W. S., Yap, M. G. S., Recombinant protein

therapeutics from CHO cells - 20 years and counting. Chem Eng Prog 2007, 103, 40-47.

[28] Narva, E., Autio, R., Rahkonen, N., Kong, L. J., Harrison, N., Kitsberg, D., Borghese, L.,

Itskovitz-Eldor, J., Rasool, O., Dvorak, P., Hovatta, O., Otonkoski, T., Tuuri, T., Cui, W.,

Brüstle, O., Baker, D., Maltby, E., Moore, H. D., Benvenisty, N., Andrews, P. W., Yli-Harja,

O., Lahesmaa, R., High-resolution DNA analysis of human embryonic stem cell lines reveals

culture-induced copy number changes and loss of heterozygosity. Nature Biotechnology 2010,

28, 371-U103.

Page 24: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

24

[29] Laurent, L. C., Ulitsky, I., Slavin, I., Tran, H., Schork, A., Morey, R., Lynch, C.,

Harness, J. V., Lee, S., Barrero, M. J., Ku, S., Martynova, M., Semechkin, R., Galat, V.,

Gottesfeld, J., Izpisua Belmonte, J. C., Murry, C., Keirstead, H. S., Park, H. S., Schmidt, U.,

Laslett, A. L., Muller, F. J., Nievergelt, C. M., Shamir, R., Loring, J. F., Dynamic Changes in

the Copy Number of Pluripotency and Cell Proliferation Genes in Human ESCs and iPSCs

during Reprogramming and Time in Culture. Cell Stem Cell 2011, 8, 106-118.

[30] Vishwanathan, N., Le, H., Jacob, N. M., Tsao, Y. S., Ng, S. W., Loo, B., Liu, Z.,

Kantardjieff, A., Hu, W. S., Transcriptome dynamics of transgene amplification in Chinese

hamster ovary cells. Biotechnol Bioeng 2014, 111, 518-528.

[31] Kantardjieff, A., Jacob, N. M., Yee, J. C., Epstein, E., Kok, Y. J., Philp, R., Betenbaugh,

M., Hu, W. S., Transcriptome and proteome analysis of Chinese hamster ovary cells under

low temperature and butyrate treatment. J Biotechnol 2010, 145, 143-159.

[32] Lee, C. J., Seth, G., Tsukuda, J., Hamilton, R. W., A Clone Screening Method Using

mRNA Levels to Determine Specific Productivity and Product Quality for Monoclonal

Antibodies. Biotechnol Bioeng 2009, 102, 1107-1118.

[33] Reisinger, H., Steinfellner, W., Stern, B., Katinger, H., Kunert, R., The absence of effect

of gene copy number and mRNA level on the amount of mAb secretion from mammalian

cells. Appl Microbiol Biot 2008, 81, 701-710.

[34] Tsuruta, L. R., dos Santos, M. L., Yeda, F. P., Okamoto, O. K., Moro, A. M., Genetic

analyses of Per.C6 cell clones producing a therapeutic monoclonal antibody regarding

productivity and long-term stability. Appl Microbiol Biot 2016, 100, 10031-10041.

[35] Hu, Z., Guo, D., Yip, S. S., Zhan, D., Misaghi, S., Joly, J.C., Snedecor, B. R., Shen, A.

Y., Chinese hamster ovary K1 host cell enables stable cell line development for antibody

molecules which are difficult to express in DUXB11-derived dihydrofolate reductase

deficient host cell. Biotechnol Prog 2013, 29, 980-985.

[36] Reinhart, D., Sommeregger, W., Debreczeny, M., Gludovacz, E., Kunert, R., In search of

expression bottlenecks in recombinant CHO cell lines-a case study. Appl Microbiol

Biotechnol 2014.

Page 25: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

25

[37] Ellgaard, L., Helenius, A., Quality control in the endoplasmic reticulum. Nat Rev Mol

Cell Biol 2003, 4, 181-191.

[38] Simmen, T., Lynes, E. M., Gesson, K., Thomas, G., Oxidative protein folding in the

endoplasmic reticulum: tight links to the mitochondria-associated membrane (MAM).

Biochim Biophys Acta 2010, 1798, 1465-1473.

[39] Kunert, R., Reinhart, D., Advances in recombinant antibody manufacturing. Appl

Microbiol Biotechnol 2016, 100, 3451-3461.

[40] Liu, L., Antibody glycosylation and its impact on the pharmacokinetics and

pharmacodynamics of monoclonal antibodies and Fc-fusion proteins. J Pharm Sci 2015, 104,

1866-1884.

[41] Chung, S., Quarmby, V., Gao, X., Ying, Y., Lin, L., Reed, C., Fong, C., Lau, W., Qiu, Z.

J., Shen, A., Vanderlaan, M., Song, A., Quantitative evaluation of fucose reducing effects in a

humanized antibody on Fcγ receptor binding and antibody-dependent cell-mediated

cytotoxicity activities. MAbs 2012, 4, 326-340.

[42] Yu, M., Brown, D., Reed, C., Chung, S., Lutman, J., Stefanich, E., Wong, A., Stephan, J.

P., Bayer, R., Production, characterization, and pharmacokinetic properties of antibodies with

N-linked mannose-5 glycans. MAbs 2012, 4, 475-487.

[43] Jefferis, R., Glycosylation as a strategy to improve antibody-based therapeutics. Nat Rev

Drug Discov 2009, 8, 226-234.

[44] Gramer, M. J., Eckblad, J. J., Donahue, R., Brown, J., Shultz, C., Vickerman, K., Priem,

P., van den Bremer, E. T., Gerritsen, J., van Berkel, P. H., Modulation of Antibody

Galactosylation Through Feeding of Uridine, Manganese Chloride, and Galactose. Biotechnol

Bioeng 2011, 108, 1591-1602.

[45] Grainger, R. K., James, D. C., CHO Cell Line Specific Prediction and Control of

Recombinant Monoclonal Antibody N-Glycosylation. Biotechnol Bioeng 2013, 110, 2970-

2983.

[46] Morell, A. G., Gregoriadis, G., Scheinberg, I. H., Hickman, J., Ashwell, G., The role of

sialic acid in determining the survival of glycoproteins in the circulation. J Biol Chem 1971,

246, 1461-1467.

Page 26: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

26

[47] Li, T., DiLillo, D. J., Bournazos, S., Giddens, J. P., Ravetch, J. V., Wang, L. X.,

Modulating IgG effector function by Fc glycan engineering. Proc Natl Acad Sci U S A 2017,

114, 3485-3490.

[48] Buchanan, A., Clementel, V., Woods, R., Harn, N., Bowen, M. A., Mo, W., Popovic, B.,

Bishop, S. M., Dall'Acqua, W., Minter, R., Jermutus, L., Bedian, V., Engineering a

therapeutic IgG molecule to address cysteinylation, aggregation and enhance thermal stability

and expression. MAbs 2013, 5, 255-262.

[49] Garber, E., Demarest, S. J., A broad range of Fab stabilities within a host of therapeutic

IgGs. Biochem Biophys Res Commun 2007, 355, 751-757.

[50] Shen, Y., Zeng, L., Zhu, A., Blanc, T., Patel, D., Pennello, A., Bari, A., Ng, S., Persaud,

K., Kang, Y. K., Balderes, P., Surguladze, D., Hindi, S., Zhou, Q., Ludwig, D. L., Snavely,

M., Removal of a C-terminal serine residue proximal to the inter-chain disulfide bond of a

human IgG1 lambda light chain mediates enhanced antibody stability and antibody dependent

cell-mediated cytotoxicity. MAbs 2013, 5, 418-431.

Page 27: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

27

9. List of table captions

Table 1. Primer sequences and qPCR settings. PCR was performed as 2-step protocol with an

initial denaturation step at 95 °C for 3 min followed by 40 cycles of annealing and extension

at described conditions and denaturation at 95 °C for 15 s.

Target Primer sense Primer antisense Amplicon

size [bp]

Annealing,

extension

PG9 HC CTGCAACGTGAATCACAAGC GTGGGCATGTGTGAGTTTTG 86 65°C, 60 s

PG9 LC ACCACACCCTCCAAACAAAG ACCTGGCAGCTGTAGCTTTT 101 65°C, 60 s

EIF3I TGGCCCTACTGAAGACCAAC CACTGGTACCCCATCTGCTT 110 64°C, 90 s

ACTB GATATCGCTGCGCTCGTT CCACGATGGATGGGAAGAC 97 60°C, 60 s

Page 28: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

28

Table 2. Summary of process relevant data of CHO-K1, CHO-S and CHO-DG44 batch, fed-batch and semi-continuous perfusion cultures based on

ActiCHO P or CD CHO media. Batch and fed-batch values represent the mean of three replicate experiments, or the mean values after day 3

(perfusion cultures) including one standard deviation.

Cell line CHO K1 CHO S CHO DG44

Medium ActiCHO P CD CHO ActiCHO P CD CHO ActiCHO P CD CHO

Peak cell concentration

(106 cells/mL)

Peak mAb

(mg/L)

Specific growth rate: day 0-5

(d-1)

Specific mAb productivity

(pg/cell/day)

Space time yield

(mg/L/d)

Batch

Fed-batch

Perfusion

Batch

Fed-batch

Perfusion

Batch

Fed-batch

Perfusion

Batch

Fed-batch

Perfusion

Batch

Fed-batch

Perfusion

7.2 (±1.0)

15.7 (±0.9)

25.4

390 (±35)

460 (±20)

365

0.56 (±0.01)

0.61 (±0.01)

n.a.

12.0 (±1.7)

6.9 (±0.4)

12.9 (±3.1)

39 (±4)

42 (±2)

301

5.0 (±0.2)

3.5 (±0.2)

25.0

300 (±5)

350 (±20)

370

0.61 (±0.00)

0.48 (±0.01)

n.a.

14.0 (±0.6)

13.8 (±0.5)

16.2 (±2.8)

38 (±1)

29 (±1)

276

11.4 (±0.8)

13.8 (±2.5)

48.2

55 (±5)

370 (±15)

85

0.76 (±0.01)

0.70 (±0.02)

n.a.

1.9 (±0.2)

3.0 (±0.2)

1.6 (±0.5)

8 (±1)

23 (±1)

48

5.9 (±0.5)

6.6 (±0.7)

41.4

75 (±1)

175 (±5)

95

0.73 (±0.02)

0.66 (±0.02)

n.a.

3.2 (±0.1)

4.2 (±0.2)

2.4 (±0.5)

9 (±0)

16 (±1)

76

10.8 (±1.6)

24.1 (±1.1)

66.0

125 (±5)

670 (±20)

345

0.59 (±0.03)

0.59 (±0.02)

n.a.

2.6 (±0.1)

5.7 (±0.1)

4.8 (±1.1)

12 (±1)

56 (±1)

237

8.9 (±0.9)

9.1 (±0.7)

27.1

105 (±5)

115 (±5)

110

0.64 (±0.01)

0.59 (±0.01)

n.a.

2.5 (±0.3)

2.3 (±0.1)

4.0 (±0.9)

11 (±1)

10 (±1)

81

Page 29: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

29

10. Figure legends

Figure 1. Total cell concentrations and viable cumulative cell days (VCCD) versus mAb

concentration of CHO-K1, CHO-S and CHO-DG44 batch, fed-batch and semi-continuous

perfusion cultures in ActiCHO P or CD CHO media. Batch and fed-batch values represent the

mean of three replicate experiments. Error bars show one standard deviation. Perfusion

cultures were operated as single runs, whereas each day represents a replicate. For perfusion

cultures the cumulative mAb titer is shown.

Figure 2. Mean values for (A) heavy chain mRNA, (B) light chain mRNA and (C) cell-

specific mAb production rates during fed-batch cultivation of CHO-K1, CHO-S and CHO-

DG44 cells in ActiCHO P or CD CHO media on days 4, 7 and the day of culture termination

(criterion = viability < 60%). All values represent the mean of three replicate experiments

with error bars showing one standard deviation.

Figure 3. Glycoform distribution of heavy chain of mAb expressed in CHO-K1, CHO-S and

CHO-DG44 fed-batch cultures based on ActiCHO P or CD CHO media. Antibody quality of

(A) G0F, (B) G1F, (C) G2F, (D) G0F-Gn, (E) G0, (F) Man5, (G) non-glycosylated and (H)

total percentage of galactosylated mAb was analyzed of the harvest product fraction after fed-

batch termination. All values represent the mean of three replicate experiments, the error bars

show one standard deviation. Statistical analysis was performed by two-way analysis of

variance (ANOVA) with post-hoc Bonferroni testing (n.s., non-significant, p > 0.05;* p≤

0.05; ** p ≤ 0.01; *** p ≤ 0.001; **** p ≤ 0.0001).

Figure 4. Glycoform distribution of light chain of mAb expressed in CHO-K1, CHO-S and

CHO-DG44 fed-batch cultures based on ActiCHO P or CD CHO media. Antibody quality

Antibody quality of (A) G0F, (B) G1F, (C) G2F, (D) G0F-Gn, (E) G0, (F) Man5, (G) total

percentage of galactosylated mAb, (H) G1F+SA1, (I) G2F+SA1, (J) G2F+SA2 was analyzed

Page 30: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

30

of the harvest product fraction after fed-batch termination. All values represent the mean of

three replicate experiments, the error bars show one standard deviation. Statistical analysis

was performed by two-way analysis of variance (ANOVA) with post-hoc Bonferroni testing

(n.s., non-significant, p > 0.05;* p≤ 0.05; ** p ≤ 0.01; *** p ≤ 0.001; **** p ≤ 0.0001).

Figure 5. Thermal stability of mAb expressed in CHO-K1, CHO-S and CHO-DG44 fed-batch

cultures based on ActiCHO P or CD CHO media. Antibody quality was analyzed of the

harvest product fraction after fed-batch termination. All values represent the mean of three

replicate experiments, the error bars show one standard deviation. Statistical analysis was

performed by two-way analysis of variance (ANOVA) with post-hoc Bonferroni testing (n.s.,

non-significant, p > 0.05;* p≤ 0.05; ** p ≤ 0.01; *** p ≤ 0.001; **** p ≤ 0.0001).

Page 31: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

31

Figure 1

Page 32: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

32

Figure 2

Page 33: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

33

Figure 3

Page 34: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

34

Figure 4

Page 35: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

 

35

Figure 5

Page 36: Bioprocessing of recombinant CHO-K1, CHO-DG44 and CHO ...download.xuebalib.com/z191xlnezTU.pdfCHO-DG44 host cell lines are deficient in the dhfr gene, which is typically co-transfected

本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

提供一站式文献检索和下载服务”的24 小时在线不限IP

图书馆。

图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

图书馆导航:

图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具