You are on page 1of 28

Optimization of Protein Expression in

Mammalian Cells
Molly Hunter,1 Ping Yuan,1 Divya Vavilala,1 and Mark Fox1,2
1
ATUM, Newark, California
2
Corresponding author: mfox@atum.bio

Recombinant proteins, such as monoclonal antibodies, are produced in mam-


malian cell lines to introduce proper protein folding and post-translational
modifications, which are essential for full biological activity. In both the indus-
trial and academic environments, the use of recombinant proteins varies widely
and, with it, the method of production. The amount of an antibody needed for
a toxicity study is far greater than that needed by a research lab performing
cellular assays, and the amount of effort put into the development of the protein
will vary accordingly. There is no universal strategy for mammalian expression
systems, and scientists often struggle to develop a suitable process from the
myriad of choices at each step. Here, we elaborate on the various obstacles en-
countered when planning high-yield experiments to produce the recombinant
proteins of interest. 
C 2018 by John Wiley & Sons, Inc.

Keywords: expression vector r gene optimization r mammalian cell culture


r protein expression r recombinant protein r stable cell lines r transient
expression

How to cite this article:


Hunter, M., Yuan, P., Vavilala, D., & Fox, M. (2018). Optimization of
protein expression in mammalian cells. Current Protocols in Protein
Science, e77. doi: 10.1002/cpps.77

INTRODUCTION scale (De Jesus & Wurm, 2011). Many of these


Mammalian protein production has become developments are also available to scientists.
standard for the manufacture of state-of-the-art Despite these advancements, selecting a pro-
biotherapeutics, as well as becoming routine in cess for recombinant protein expression is not
the production of protein samples for research. necessarily simple. Variables such as cell line,
Protein production in mammalian cell lines culture media, and culture method all impact
offers the unique advantage of generating as the yield, physical characteristics, and biologic
close to wild-type environments for transcrip- activity of the proteins expressed, as depicted
tion and translation, coupled with the relevant in Figure 1. This review focuses on the prac-
chaperone, secretory, and redox environments tical aspects of developing a strategy for the
and post-translational modifications that lead rapid, economical production of proteins in
to functionally relevant and active proteins. mammalian hosts that will be of use to new
While it is possible to produce human proteins scientists.
in prokaryotic expression systems, the result-
ing proteins often have undesirable character- CELL LINES AS EXPRESSION
istics that impact efficacy, such as a lack of HOSTS
glycosylation. Currently, prokaryotic systems The most commonly used mammalian cell
cannot match the advantages of mammalian lines found in research and industrial set-
systems. Much of the technological advance- tings for protein production are Chinese ham-
ment in mammalian systems has been focused ster ovary cells (CHO) and human embry-
on industrial processes, especially the produc- onic kidney 293 cells (HEK-293) (S. Estes &
tion of biotherapeutics at the multi-kilogram Melville, 2014; Hacker et al., 2013; Swiech,

Hunter et al.

Current Protocols in Protein Science e77 1 of 28


Published in Wiley Online Library (wileyonlinelibrary.com).
doi: 10.1002/cpps.77

C 2018 John Wiley & Sons, Inc.
Figure 1 A summary of the many elements that need to be considered for the development of
a rapid and economical mammalian expression system. Optimization of these factors cannot be
made in isolation; gene optimization, vector selection, type of selection pressure, molecule type
to be expressed, transient or stable expression, cell type, media selection, production scale, and
post-translational modifications all affect the final protein yield and biological activity, and they are
interdependent.

Picanco-Castro, & Covas, 2012). Hundreds cell lines can produce and post-translationally
of other cell lines exist, and a considerable modify eukaryotic proteins in functional
amount of effort has been employed in the en- forms at high levels. HEK-293 and CHO cells
gineering of the next generation of high pro- in suspension typically have a doubling time
ducing cell lines, but HEK-293 and CHO are of less than 24 hours and can grow to densities
the mainstays. The CHO cell line was culti- higher than 5 million cells per ml, depending
vated from a biopsy of the ovary of the Chi- on culture conditions. Both cell types are
nese hamster. The HEK-293 cell line was cre- readily transfected or virally transduced to
ated when human embryonic kidney cells were introduce foreign DNA that codes for target
transformed with fragments of adenovirus type proteins. Productivities of grams of protein per
5 DNA, resulting in a cell line that replicated liter of culture, generated by transient trans-
rapidly, tolerated reduced serum concentra- fection, have been reported for both HEK-293
tions in the media, and was easily transformed and CHO cells. Higher yields are possible
(Graham, Smiley, Russell, & Nairn, 1977). from stable pools, and clonally selected stable
Both HEK-293 and CHO cells have since been lines have been reported expressing tens of
genetically modified to improve responsive- grams per liter, with cellular productivities of
ness to different expression vector elements. more than 50 pg/cell/day (Huang et al., 2010;
For example, the HEK-293T derivative en- C. Liu, Dalby, Chen, Kilzer, & Chiou, 2008).
codes the SV40 Large T antigen, allowing HEK-293 and CHO lines are available
for episomal replication of plasmids contain- from cell line repositories, such as the
ing the SV40 origin of replication (DuBridge American Type Culture Collection (ATCC;
et al., 1987; Rio, Clark, & Tjian, 1985). The Rockville, MD), or through commercial
CHO-K1 derivative was cultured from a clone suppliers. Genetically modified cell lines, in
of the CHO line and contains fewer chromo- which key steps of a metabolic pathway are
somes than the original (Kao & Puck, 1968). mutated or removed, are also available, and
HEK-293 and CHO cells are maintained they are vital for selection procedures used in
as either suspension or adherent cultures, with the generation of stable cell lines. Suspension
suspension culture being particularly favored cell kit-based systems that package optimized
Hunter et al. for their ease of handling and scale up. Both cell lines, media, transfection reagents, and
2 of 28
Current Protocols in Protein Science
their corresponding protocols are convenient, to attach core fucose to nascent N-glycan moi-
work well, and are accessible to anyone (Taki eties include suppression of the three key genes
et al., 2015). Examples include the Expi involved in fucose modification using RNAi
systems from ThermoFisher Scientific, the (Imai-Nishiya et al., 2007), overexpression
MEXi system from IBA Lifesciences, and of b(1,4)-N-acetylglucosaminyltransferase III
the Epi-CHO system from Acyte Biotech (GnTIII) (Umana, Jean-Mairet, Moudry, Am-
(C. Liu et al., 2008; Prevato et al., 2015; Taki stutz, & Bailey, 1999), and bacterial oxidore-
et al., 2015). Even though HEK-293 and CHO ductase GDP-6-deoxy-D-lyxo-4-hexulosa re-
are most frequently used, other cell lines are ductase (RMD) (von Horsten et al., 2010). The
available for protein production. The NS0 challenge with these processes is to prevent
and Sp2/0 lines were derived from mice and significant changes in cell character beyond
used to generate drugs such as Synagis and fucosylation. High-mannose cell lines have
Erbitux (Butler & Spearman, 2014; Chartrain been introduced in recent years, created ei-
& Chu, 2008; Johnson et al., 1997; Peakman ther through mutagenesis and selection pres-
et al., 1994). New lines such as the CAP and sure (Stanley, 1983) or by site-directed mu-
CAP-T, from human amniocyte (Fischer et al., tagenesis of the Mgati gene (Sealover et al.,
2012), and PER.C6 R
(Jones et al., 2003), 2013). Enhanced ADCC activity of antibod-
from human retina, are reported to have ies with high mannose glycans is well docu-
very high cellular productivities and generate mented, but they are also known to decrease
human-type post-translational modifications. serum half-life (Reusch & Tejada, 2015). The
Scientists are continually researching and sialylation of a glycoprotein has also been
engineering new cell lines with desired at- found to play a significant role in serum half-
tributes, such as higher yields, specific post- life. Cell engineering efforts have identified
translational modifications, or improved se- various gene targets for enhancing sialic acid
lection pressure for stable cell lines, among biosynthesis, transfer, and preservation with
others. The availability of omics data has the goal of improving serum half-life. Current
dramatically improved our understanding of approaches include the overexpression of ei-
production cell lines, allowing cell line im- ther UDP-GlcNAc2-epimerase or CMP-sialic
provements to be made via cell line engineer- acid transporter, RNA interference of siali-
ing instead of random mutagenesis (Xu et al., dases, and the overexpression of sialyltrans-
2011). The discoveries of the CRISPR/Cas9 ferases such as St6gal (Lin et al., 2015). In all
(Cong et al., 2013; Jinek et al., 2013; Mali of these cases, glycosylation modulation has
et al., 2013), zinc finger nucleases (Carroll, been shown to have a significant impact on
2011), and transposases (Mates et al., 2009) clinical efficacy and pharmacokinetics.
systems have accelerated the development of Despite the variety of mammalian cell lines
cell line engineering technologies. Of partic- available for protein production, CHO cells
ular interest are the glycosylation profiles of remain the workhorse of biopharmaceutical
secreted proteins and their effect on func- production since the approval of the first CHO-
tion. For antibodies, glycosylation can dic- derived recombinant protein, tissue plasmino-
tate properties such as antibody-dependent gen activator, tPA, in 1986. The process of
cellular cytotoxicity (ADCC), complement - biomanufacturing in CHO cells involves cell
dependent cytotoxicity (CDC), and serum line development and screening to select the
half-life. Changes to the glycosylation pro- stable pool or clone that has the highest cell vi-
file of a protein can be accomplished either ability, growth, expression level, and product
by modifying the genome of the cell or by quality. CHO cells are involved in the produc-
co-expression of plasmids encoding other fac- tion of over 70% of therapeutic recombinant
tors (Le Fourn, Girod, Buceta, Regamey, & proteins, most of which are monoclonal anti-
Mermod, 2014; Yin et al., 2015). The corre- bodies (Jayapal, Wlaschin, Hu, & Yap, 2007).
lation between low levels of core-fucosylation In 2017, the global revenue from monoclonal
and increased ADCC activity (Shinkawa et al., antibodies was over $90 billion, providing a
2003) has led to the creation of cell lines strong incentive for the continued develop-
with reduced or eliminated core-fucosylation ment of cell line engineering technologies.
of secreted proteins. The FUT8 knockout
CHO cell line is unable to synthesize α(1,6)-
fucosyltransferase, resulting in the expres- CELL CULTURE MEDIA FOR
sion of completely defucosylated antibodies PROTEIN PRODUCTION
(Yamane-Ohnuki et al., 2004). Methods for re- Modern cell culture media was developed Hunter et al.
moving the ability of existing stable cell lines over 60 years ago starting with 199 (M199;
3 of 28
Current Protocols in Protein Science
Morgan, Morton, & Parker, 1950) and Harry that are not only free of animal-derived compo-
Eagle’s minimum essential medium (MEM; nents but are also chemically defined, meaning
Eagle, 1959). While media formulations are that the quantity of each component is known.
tailored for specific applications, they all have Development of a sophisticated defined media
the same goal: to supply the nutrients re- is not without significant challenges. Compo-
quired for healthy cell division and propaga- nents in culture media frequently interact due
tion. Due to the ease of use, complex media to the complexity of cellular metabolic path-
components such as fetal bovine serum, serv- ways. Furthermore, nutritional requirements
ing as cellular protectants against shear, pH are defined by the system in which the cell
change, and nutrient stress were favored in is used. It cannot not be expected that media
the past for protein production. However, con- designed for transient transfection in CHO
cerns of undefined raw materials potentially cells to be the best media for clonal selection
containing adventitious agents, in conjunction of a CHO stable line. Transient expression
with serious ethical issues regarding the wel- media must contain the appropriate nutrients
fare of the donor fetus during harvest (Asher, to support the growth of high density cultures
1999) pushed the industry to replace serum expressing recombinant proteins, while media
with defined raw materials. For example, in for clonal cell line development must promote
the 1990s, bovine spongiform encephalopa- the growth of cells at extremely low densities.
thy/transmissible spongiform encephalopathy Selecting a media to produce recombinant
(BSE/TSE) spread in beef cattle in the United proteins can be as simple or complicated as
Kingdom, resulting in the deaths of over 200 the process requires. For a commercially avail-
people. As a result, the United States Food and able expression system like those offered by
Drug Administration restricted the importa- HycloneTM (GE Life Sciences) and GibcoTM
tion of bovine serum from countries that were (ThermoFisher), the choice is simple: cell lines
affected by the BSE epidemic (Asher, 1999; can be easily adapted to these commercial me-
Keen & Rapson, 1995). Cell culture media dia, or cells can be bought in a kit with the
for mammalian cell production has now pro- media and transfection reagents. However, for
gressed to formulations that are serum-free or more sophisticated processes, the decision of
chemically defined, typically containing 50 to which media to use will significantly affect
100 components grouped as energy substrates, the amount and quality of the biotherapeutics
chelators, amino acids, surfactants, pH buffers, made. High cell densities and cell viability lead
vitamins, nucleic acid derivatives, trace el- to higher protein productivities. However, the
ements, salts, fatty acids, and lipids (Keen nutritional requirements for cell growth can
& Rapson, 1995; McCoy, Costa, & Morris, conflict with those for production of proteins.
2015). The added benefit of serum-free cell Media composition is divided between these
culture media is that it simplifies the down- two tasks. An initial approach would be to
stream purification process. assess the qualities of serum-free and chemi-
Published literature is sparse as to the cally defined media sold by a variety of man-
exact composition of many of the latest media ufacturers. Cells may require a series of adap-
formulations, as it is proprietary information tation steps to new growth media, typically
for commercial products readily available performed by weaning, in which the new and
from suppliers such as HycloneTM (GE Life old media are mixed, gradually increasing the
Sciences), GibcoTM (Thermo Fisher Scien- proportion of new media as the cells adapt.
tific), Lonza and Irvine Scientific. Serum-free Changing the media may lead to improved cell
medium consists of nutritionally complete growth and productivity but must not come at
basal medium supplemented with an empiri- the expense of an unacceptable change in pro-
cally determined mixture of hormones, growth tein quality, such as glycosylation, proteolytic
factors, attachments factors, and binding pro- cleavage, or protein aggregation.
teins. These mixtures are commercially avail- Component titration is the classic approach
able and referred to as serum replacements to media development, involving a series of
(Keen & Rapson, 1995). Vegetable-derived experiments to determine the cell line’s dose
protein hydrolysates have been shown to be response to various media components, adding
a capable replacement for serum and animal- each one at a time. For example, there have
derived components (Pazos et al., 2004). How- been several reports on the specific effects of
ever, hydrolysates by their nature are complex copper in cell culture application. In one study
and not fully defined. The latest step in media of CHO cells expressing the IgG-fusion pro-
technology has been to develop culture media tein B0, the addition of 50 μM copper sulfate
Hunter et al.

4 of 28
Current Protocols in Protein Science
to the media reduced lactate and increased cell variety of cell feeds and hydrolysates are avail-
viability and protein yield. However, reducing able that contain glucose, amino acids, lipids
sulfate concentration to 5 μM had the same and other factors to maintain the culture (Kyr-
positive effects, as well as reduced protein iakopoulos & Kontoravdi, 2014; McCoy et al.,
aggregation (Qian et al., 2011). Copper sul- 2015; Mosser et al., 2013). DOE can be used to
fate has also been shown to increase the pro- screen and optimize feed types, amounts, and
ductivity of recombinant factor VIII expressed timing. Cell culture metabolite data that are
in both baby hamster kidney cells (BHK) and measured by a Nova system, or more simply
CHO cells (Chan & Harris, 1997). Component with glucose testing kits, can also be applied
titration is labor-intensive (a typical cell cul- to the DOE. It must be noted that glucose feed-
ture media contains 50 to 100 ingredients) and ing can lead to harmful lactate buildup, which
does not always reveal interactions between may need to be monitored along with increas-
components. Media blending is an alternative ing ammonium levels, to avoid inhibiting pro-
method that rapidly generates new media by tein production and altering glycosylation (Y.
mixing existing formulations. An excellent ex- Fan et al., 2015; B. Liu et al., 2014). Feeding
ample of such an approach is the blending of and media additions must not change the cell
the classical formulations DMEM and Ham’s culture osmolality significantly, as this may
F12 media, resulting in DMEM/F12, a ubiq- also affect productivities and protein quality.
uitous formulation that is frequently used as Commercial cell culture media typically have
a basal media (D. Barnes & Sato, 1979). Al- buffering systems, such as HEPES, that work
though blending is an effective and faster pro- alongside bicarbonate buffering provided in
cess, the performance of such mixtures can- conjunction with CO2 from the cell culture in-
not be attributed to a single component within cubator (Zigler, Lepe-Zuniga, Vistica, & Gery,
the media, making troubleshooting difficult. 1985). pH is also actively controlled in biore-
Nevertheless, this approach can be very suc- actors if this equipment is available (Howorth,
cessful when mixing media that differ only 1975).
by a few elements. It is possible to increase One of the most critical parameters to
growth and productivity by following a statis- consider when developing cell culture me-
tical approach to media mixing using a design dia is the method of transfection to be used.
of experiments (DOE) methodology (Mande- There are several commercially available for-
nius & Brundin, 2008). Spent media analy- mulations that can be used with both HEK-
sis provides a stoichiometric approach to me- 293 and CHO cells that allow levels of re-
dia design. Analysis of nutrient consumption combinant protein expression in the range of
can deliver information regarding nutritional many hundreds of milligrams per liter. How-
requirements and culture chemistry changes ever, several contain ingredients that can in-
over the course of a process, providing insights hibit transfection. Iron (III) citrate (Eberhardy,
into the balance of amino acids and other nu- Radzniak, & Liu, 2009), and dextran sulfate
trients. However, assays for every media com- both inhibit polyethylenimine-mediated trans-
ponent are not available. Most assays are lim- fection but can improve protein production
ited to glucose, lactate, ammonia, and amino if added later in the process. Other compo-
acids, delivering a simplistic overview of a far nents may increase protein production lev-
more complex biological problem. Ultimately, els by aiding transient transfection efficien-
rational culture media design requires that all cies, such as N,N dimethylacetamide (DMA;
approaches are considered and used when ap- Rajendra et al., 2015), lithium acetate (LiAc;
propriate (Fletcher, 2005). Ye et al., 2009) and dimethyl sulfoxide
Nutritional considerations for stable cell (DMSO; Ye et al., 2009). Further enhancement
fed-batch systems differ from transient sys- of protein expression for both transient and
tems in that the media for fed-batch cultures stable line producers is achieved by supple-
must support cell growth and production of menting the expression phase of a process with
the product for much longer time periods than small molecule additions. In particular, the
transient cultures. For secreted proteins and use of histone deacetylase inhibitors, such as
cytoplasmic proteins, titers are proportional to sodium butyrate (Mimura et al., 2001) and val-
cell density, so in batch culture, generating and proic acid, have been shown to increase protein
maintaining high viability, high-density cul- titers in both HEK-293 and CHO cells (Backli-
ture is crucial. At high cell densities, nutrients wal et al., 2008). Most of these inhibitors mod-
are rapidly depleted. Glucose feeding is rou- ify host histone acetylation or methylation pat-
tinely employed to support cell growth, and a terns, thereby increasing cellular transcription
Hunter et al.

5 of 28
Current Protocols in Protein Science
levels and boosting protein production many made from mammalian cells, is still produced
folds. The concentration and timing of addi- in adherent CHO cells cultivated in roller bot-
tions must be determined empirically as they tles (F. M. Wurm, 2004). Although much of
may result in cellular toxicity and arrest of cell the process has been roboticized, the classic
growth (Backliwal et al., 2008). Other small roller bottle remains the pillar of the process
molecules have been shown to have positive (F. M. Wurm, 2004).
impacts on protein production, such as LiAc
(Cervera et al., 2015) and caffeine (Cervera Traditional Methods
et al., 2015; Dell, Mcgrew, Trentalange, & The challenge of scaling traditional ad-
Van, 2004). herent flask-based processes is addressed by
Finally, media composition may also af- maximizing the total surface area in a planar
fect post-translational modifications of pro- vessel containing multiple layers. Multilayer
teins. Of these modifications, glycosylation vessels have been available for over 30 years
has been the most extensively studied due to its (Stacey & Davis, 2007) and are commercially
ability to influence drug clearance rates, spe- available as stacked-plate systems called
cific activity, solubility, immunogenicity, and Cell FactoriesTM (NuncTM ) and CellStacks R

R
further processing of the protein. Various nu- (Corning ). Stacked-plate systems have
trients, sugar precursors, amino acids, trace allowed a traditional T-25 laboratory flask,
metals, and hormones have been shown to in- with a surface area of 25 cm2 , to be scaled to
fluence glycan distribution. Glucose starvation 6,360 cm2 in a 10-layered vessel, 25,440
has been shown to reduce the incidence of gly- cm2 in a 40-layer vessel, and 60,000 cm2
cosylation at asparagine residues, while the in a 120-layer hyperstack R
vessel (Stacey &
supplementation of the sugar galactose in the Davis, 2007). Increased surface area brings
presence of manganese chloride (MnCl2 ) can its own set of challenges, including even
alter the types and abundance of the glycosy- distribution of nutrients, gas exchange, and
lation (St Amand, Radhakrishnan, Robinson, the manual manipulation of the stacks. Equip-
& Ogunnaike, 2014). Controlling osmolality ment manufacturers have addressed these
of the cell culture has been shown to provide issues by adding bioreactor control to planar
a means to control fucosylation during per- culture systems and creating more compact
fusion and fed-batch cultures. Hypoosmolal- systems. For example, the xPansion R
mul-
R
ity of the culture has been shown to reduce tilayer bioreactor (PALL ) provides a cell
levels of fucosylation of monoclonal antibod- growth surface area up to 122,400 cm2 with
ies (Konno et al., 2012). The impact of me- temperature monitoring, agitation control, and
dia composition changes on post-translational disposable pH and dissolved oxygen sensors.
modifications must be taken into consideration Researchers needing to scale up adher-
during development, as it can have a direct ent expression systems for secreted proteins
impact on the biological activity of the target should also consider a process based on mi-
protein. crocarriers, which can be applied to stir tanks
(Hu et al., 2000) and wave bioreactors (Singh,
1999). This approach allows for optimization,
CULTURE METHODS flexibility in feeding parameters, scalability
Traditional laboratory-scale tissue culture up to 10,000 liters, and a higher surface area
using culture dishes seldom produces more to volume ratio. A less common combina-
than a few micrograms of purified recombinant tion of bioreactors and adherent cultures is the
proteins, antibodies, or viruses from cultures packed-bed bioreactor, in which the cells are
of adherent cells. With burgeoning needs for immobilized on a substrate that is enclosed
milligrams of protein for crystallography, drug in a packed bed within a bioreactor. A wide
discovery, or feasibility tests of a novel drug, array of substrates and configurations exist,
the static tissue culture dish has been replaced but the most widely used are beads, porous
by large-scale systems. Cell culture scale-up structures, fibers and hollow fibers (Meuwly,
of adherent cells has become routine since the Ruffieux, Kadouri, & von Stockar, 2007; Park
development of the round glass culture dish & Stephanopoulos, 1993; Wang et al., 1992).
by Richard Petri in 1877, cell culture flasks The primary advantage of a packed-bed cul-
developed by Alex Carrel in 1923, and roller ture is the lower shear-stress forces on the
bottles, the concept of growing cells as rotat- cells, compared with microcarriers that are ag-
ing cultures, designed by George Gey in 1933. itated in a stir tank (Brindley et al., 2011).
Some of these systems remain relevant today. Researchers who need more than a few
Hunter et al.
Erythropoeitin, the first recombinant protein milligrams of a product are encouraged to
6 of 28
Current Protocols in Protein Science
switch to a suspension-adapted cell line when and offer control of temperature, oxygenation,
possible. Suspension culture of mammalian gas exchange, pH, and other factors (Singh,
cells is operationally more facile than adherent 1999). Cell growth and product generation are
culture and is adaptable to automated liquid highly dependent on wave development and
handling and scale up to bioreactor culture propagation, which is mainly influenced by
(Muller, Girard, Hacker, Jordan, & Wurm, rock rate, rock angle, fill level, bag geom-
2005). Switching to a suspension-adapted etry, and culture media (Eibl, Loffelholz, &
cell line can be as simple as placing a small Eibl, 2014). Wave-mixed bioreactors are scal-
platform shaker and a tray of deionized H2 O, able up to 500 liters and enable rapid gener-
for humidity, inside an existing CO2 incubator. ation of material for early drug development
Tubes containing just a few milliliters of cul- either by large-scale transient expression, or
ture can be scaled up to large flasks containing expansion of stable pools or clones. Despite
multiple liters. If the goal is to test many prod- the same working principle of wave bioreac-
ucts or expression conditions, investment in a tor systems, commercial reactors differ in their
CO2 shaker incubator, available from suppli- control unit, platform movement, bag design,
ers such as Infors AG and Kuhner Shaker Inc, number of sensors, and the PID loops present
provides a unified platform from screening in each unit. Wave Rocker (GE Life Sciences),
to small-scale biomanufacturing. Variable or- BIOSTAT R
RM (Sartorius), CELL-tainer R

TM
bital shaking diameters of 3, 12.5, 25, and 50 (Biotech BV) and Allegro XRS 25 biore-
mm provide the tools to handle a wide range actor System (PALL) are commercially avail-
of cell culture volumes and containers. For able wave-mixed bioreactors. As a less expen-
high throughput testing, 96- or 24-well blocks sive option, Kuhner Shaker, Inc and Infors AG
are particularly useful for screening both supply trays that can be inserted into a CO2
transiently transfected HEK-293 cells during shaking incubator to provide an alternative to
protein engineering projects (Bos et al., 2015; an independent wave unit.
Davies, Greene, Lullau, & Abbott, 2005) and
stable CHO cells for clone selection. Also, 96- Bioreactors
and 24-deep well plates significantly increase Stir tank bioreactors are the most com-
throughput as liquid handling robots are suit- monly used bioreactor type for cultivating
able for incorporation at all stages of the pro- suspension cells. Their general design cri-
cess, including purification (Chaturvedi, Sun, teria was derived from E.coli fermentation.
O’Brien, Liu, & Brooks, 2014). As more pro- They have been successfully used for the cul-
tein is required, convenient 50 ml Tubespin R
tivation of a variety of suspension cells in-
bioreactors are available for growth at larger cluding CHO, baby hamster kidney (BHK),
scales (10 to 30 ml), and these vessels are also HEK-293, and PER.C6 R
, and are available
liquid handling compatible (Gomez et al., for culture volumes from 10 ml (Ambr R
sys-
2017; Legmann et al., 2009). Shake flasks are tem) to many thousands of liters (Janakiraman,
typically used for volumes between 30 ml and Kwiatkowski, Kshirsagar, Ryll, & Huang,
5 liters (Stettler, Zhang, Hacker, De Jesus, & 2015; Smelko et al., 2011). Stainless steel
Wurm, 2007). It may be necessary to test the tanks are used at laboratory and pilot scales
scalability of the product from the small to for research grade material but require sterility,
large scale, as differences in cell growth, shear cleaning, and maintenance. Single-use biore-
factor, oxygenation, and other factors may actors enable fast setup and breakdown and
affect protein titer and quality. It is essential to reduce contamination risks (Irfan, Rees, Bar-
use the correct shaker orbit relative to culture rett, Markicevic, & Segarra, 2015). Specific
vessel size, fill volume, and shake speed. challenges should be taken into consideration
For transient transfection, these parameters when moving to an impeller driven bioreac-
must also be considered for maintaining tor. An ideal bioreactor should impart low
the integrity of the transfection complex in shear stress on the cells, minimizing me-
culture and its ability to enter a cell during chanical damage from the eddies formed by
transfection. the impeller motion or bursting gas bubbles,
For further scale-up, wave-mixed bioreac- while ensuring uniform mixing of the nutri-
tors can be used. In wave-mixed bioreactors, ents in cell culture media and reducing gradi-
a one- or two-dimensional movement of the ents across the tank. Once optimized, stir tank
bioreactor platform induces a wave in a ster- bioreactors typically enable consistent scala-
ile bag, providing oxygen transfer and media bility and control of all culture conditions, re-
mixing throughout the culture. These bioreac- sulting in higher yields and more controllable
Hunter et al.
tors are easy to operate, minimize shear stress, product qualities.
7 of 28
Current Protocols in Protein Science
Bioreactors may be operated in batch, before committing significant amounts of time
extended batch (also known as fed-batch), and resources into stable cell line generation
or perfusion mode. Fed-batch cultivation of (Gutierrez-Granados et al., 2018).
mammalian cells is the most common, and Several different cell lines have been shown
it involves the gradual addition of selected to be capable of being transiently transfected
nutrients during the growth-culture cycle to for the expression of recombinant proteins, in-
improve productivity and growth, typically cluding HEK-293 (Jordan, Kohne, & Wurm,
lasting 10 to 14 days (Pollock, Ho, & Farid, 1998), CHO (Preuss, Connor, & Vogel, 2000),
2013). The development of an extended COS (African green monkey kidney) (Blasey,
batch process requires an understanding of Aubry, Mazzei, & Bernard, 1995) and BHK
the nutritional requirements of the cell line. (baby hamster kidney) cells (F. Wurm &
Perfusion mode enables long production runs Bernard, 1999). The HEK-293 cell line ex-
of stable cell lines (for 30 to 60 days or hibits very high transfection efficiency and
longer) as product and waste are removed and reliable translation and processing of pro-
replaced with fresh growth media (Pollock teins, resulting in higher titers compared to
et al., 2013). This culture method is particu- other mammalian cells. Significant develop-
larly amenable to the production of secreted ment work has been performed in suspen-
proteins but less so for that of cytoplasmic sion HEK-293 cells, providing many options
proteins. Perfusion growth may utilize micro- for reagents and scales for transfection (Jager
or macro-carriers that immobilize the cells, et al., 2013). Many variables, including the
protecting the cells from shear and allowing health and density of cells at transfection,
easy media exchange (Butler, Imamura, growth conditions, culture vessel, expression
Thomas, & Thilly, 1983). Other perfusion construct, duration of expression, and trans-
techniques include the use of cell separators, fection method, must be optimized to achieve
Alternating Tangential Flow (ATF)/Tangential a high level of transient expression, as protein
Flow Filtration (TFF) devices, or hollow fiber production diminishes over time due to dilu-
systems. These methods enable extremely tion of the expression vector by cell division
high live cell densities, exceeding 200 million (Hopkins, Wall, & Esposito, 2012) (Fig. 1).
cells per ml (Clincke et al., 2013). The highest The HEK-293T (Lebkowski, Clancy, & Calos,
productivity cultures reported, such as the 1985) and HEK-293EBNA (Cachianes et al.,
27 g/L antibody titer from the PER.C6 R
cells, 1993; Sun, Goh, Wong, Mori, & Yap, 2006)
typically have utilized perfusion bioreactor cell lines are integrated with a gene encoding
culture (Tchoudakova et al., 2009). simian virus 40 (SV40) large T and Epstein-
Barr nuclear antigen-1 (EBNA), respectively.
Transient vs Stable Systems Cells producing these proteins allow for epi-
The choice between transient and stable somal gene replication and segregation when
expression systems depends upon throughput, an SV40 ori or oriP element is present in the
the quantity and quality of material required, expression plasmid. This prevents dilution of
and the turnaround time. Both methods the plasmid as the cells replicate and often re-
involve getting target DNA into cells, but in sults in improved production yield (Daramola
transient protein production, the expression et al., 2014; Magistrelli et al., 2010).
vector never integrates into the genome of the One drawback of using HEK-293 expres-
cell. For large amounts of protein production, sion systems is the differeing glycosylation
stable cells are preferred, as they provide high patterns from those proteins made in CHO
yields with consistent quality. However, at cells, the ultimate host for biopharmaceuti-
the early stage of a project, where milligrams cals production. It is therefore beneficial to
to grams of protein is required, transient keep the host cell type consistent throughout
transfection is preferred, due to speed and product development, reducing the likelihood
lower cost (Gutierrez-Granados, Cervera, of changes in activity or biophysical proper-
Kamen, & Godia, 2018). Furthermore, it ties. Traditionally, CHO transient expression
provides an opportunity to focus on protein systems have been limited by transfection ef-
engineering experiments, construct design, ficiencies and titers that are insufficient for in
growth parameter improvements, and product vivo pharmacokinetic/pharmacodynamic stud-
feasibility assessments. Transient systems ies (Derouazi et al., 2004; Galbraith, Tait,
offer a product with the quality suitable Racher, Birch, & James, 2006). Both CHO
for preclinical evaluation, thus speeding the cell lines and expression vectors have been
“Proof of Concept” stage when screening engineered for improved productivity. Engi-
Hunter et al.
multiple candidates. This can be achieved neering cells for increased transcription rates
8 of 28
Current Protocols in Protein Science
and transgene expression, regulating cellu- the genome to integrate their DNA, and they
lar metabolism, reducing the induction of have inbuilt mechanisms to divert the cellu-
apoptosis, and over-expressing of XBP1 (Cain lar machinery in their favor. Lentiviral vec-
et al., 2013), ERO-La (Cain et al., 2013) or tors can transduce CHO cells with high vector
EBNA-1 and GS (Daramola et al., 2014) have copy, resulting in high levels of product that
all been shown to increase the final titer pro- is appropriately glycosylated. Integration via
duced in CHO. Engineered CHO lines such lentiviral vectors has been shown to remain
as these are generally not commercially avail- stable for many months (Baranyi et al., 2013;
able, and patents may limit their use. Com- Gaillet et al., 2010). Any virus-based protein
mercially available transient CHO expression production system raises safety issues which
systems have improved significantly, and pro- need to be addressed if the stable cell line will
tein titers observed with the ExpiCHO sys- be moved into biotherapeutic production for
tem (ThermoFisher) have been reported to be humans. An alternative to a complete viral sys-
higher than those seen in both the FreeStyle- tem is non-viral adeno-associated virus (AAV)
MAX CHO and Expi293 systems. expression. This approach takes advantage of
Stable cell lines, in particular clonally se- the AAV machinery, which integrates into spe-
lected stable cell lines, have not been sur- cific sites in the genome, to bias integration to-
passed for high levels of protein production. wards open chromatin that favors high expres-
Stable cultures can show consistent produc- sion and genomic stability (Dorai et al., 2012).
tivities for weeks, if not months, in both the Another novel viral vector system developed
batch and continuous culture. The traditional from alphaviruses, Semliki Forest virus (SFV),
strategy for making a cell line involves nu- uses an RNA vector system to generate stable
merous labor-intensive steps, from transfec- cell lines (Casales et al., 2010).
tion to the isolation of the final clonal line (Lai, Plasmids bearing genetic regulator
Yang, & Ng, 2013). The development spans elements can also be used to confer a tran-
months and consists of screening hundreds of scriptionally active state during the random
clones for productivity and quality attributes integration of a transgene into a heterochro-
before a smaller set are selected as candidate matic environment. Some of these elements,
production cell clones. Development of a re- such as scaffold/matrix attachment regions
combinant CHO cell line relies on the ran- (S/MARs) and locus control regions (LCRs),
dom integration of a plasmid into the genome function through actively switching regions of
by non-homologous recombination, an ineffi- the genome to an active transcriptional state
cient process occurring in a small portion of (Saunders, Sweeney, Antoniou, Stephens,
cells that are transfected. The cells are sub- & Cain, 2015). One of the most significant
jected to increasing levels of selective pressure challenges to reliable expression from stable
to select for transfectants that have integrated cell lines is gradual epigenetic silencing via
the gene of interest into their genome. Repeat- methylation. Another category of regulatory
ing this selection process with increased se- elements works to prevent this silencing
lection pressure can result in cell clones with a by restricting heterochromatin expansion,
high number of copies of the marker gene, co- maintaining a region of euchromatin under
amplification of the gene of interest, and high active transcription. This category includes
levels of protein expression. However, lack of insulators, ubiquitous chromatin opening ele-
control of gene insertion, as well as the effects ments (UCOEs), and stabilizing anti-repressor
of gene amplification, can lead to unwanted elements (STAR) (Neville, Orlando, Mann,
phenotypic heterogeneity due to the varying McCloskey, & Antoniou, 2017). Use of these
accessibility of integration sites. These issues plasmid elements in the creation of stable
often result in unstable cell lines that show re- CHO cells has been shown to promote protein
duced production over time (Lai et al., 2013). yields and sustain transgene expression
For long-term, consistent protein produc- (Kwaks & Otte, 2006).
tion, it is necessary to stably integrate genes Site-specific recombination can generate
of interest into the genome of the host cell high yielding stable clones reproducibly.
at sites that are transcriptionally active, that Different gene recombination systems have
do not experience gene silencing, (Wipper- been used, including the bacteriophage P1-
mann, Rupp, Brinkrolf, Hoffrogge, & Noll, derived Cre recombinase, the yeast-derived
2015) and are not susceptible to genetic re- FLP-FRT, and the C31 integrase, derived
arrangement (Dorai et al., 2012). Viral vec- from Streptomyces phage f31(Ahmadi,
tors target transcriptionally active regions of Damavandi, Akbari Eidgahi, & Davami,
Hunter et al.

9 of 28
Current Protocols in Protein Science
2016). However, these systems do require the transposase enzyme, or the mRNA coding for
establishment of a platform cell line whose the transposase. Using the purified transposase
genome is integrated either randomly or at a enzyme or its mRNA avoids overexpression
limited number of sites, with reporter cassettes of the transposase in the cell and prevents
flanked by short cis-acting DNA target se- the integration of the transposase DNA into
quences recognized by specific recombinases. the genome. Using transposases to integrate
The Cre recombinase-mediated integration the gene of interest into the genome results
process recognizes the short 34 bp cis-acting in much higher integration rates than random
DNA target sequence, loxP, and was the first integration, with fewer instances of transpo-
system to be used for human monoclonal son truncation or scrambling (Fig. 2B). Sev-
antibody production in CHO (Kawabe et al., eral approaches are commercially available,
2017). Similarly, the Flp recombinase requires including the PiggyBac Transposon system
a 48 bp Flp Recombination Target (FRT) (SBI System Biosciences), and Leap-In Trans-
sequence for recombination. Cells integrated posase system (ATUM). Both approaches have
with the FRT sequence are available from been successfully used to rapidly generate sta-
ThermoFisher, with the Flp-InTM System, and ble pools with volumetric productivities higher
have been used successfully to generate sta- than those produced by traditional random in-
bles lines that consistently produce polyclonal tegration methods (Balasubramanian, Rajen-
antibodies (Cacciatore, Leonard, & Chasin, dra, Baldi, Hacker, & Wurm, 2016; Matasci
2012; Zhou, Liu, Sun, Huang, & Yu, 2010). et al., 2011; Matasci, Baldi, Hacker, & Wurm,
The downside of both approaches is that the 2011) (Fig. 2C). The timeline for generating
site-specific recombination is reversible, as stable pools can be as little as 2 to 4 weeks
the target sequence remains after integration. (Balasubramanian et al., 2015; Balasubrama-
λ integrase and φC31 integrase target 2 dif- nian et al., 2016) from transfection to protein
ferent sequences, attP, and attB and catalyzes recovery, establishing cell pools suitable for
irreversible recombination by altering the protein production and the selection of clonal
attB and attP sites upon cassette exchange cell lines (Fig. 2D). The speed with which sta-
(Cacciatore et al., 2012). With the genome ble pools are made makes this approach a vi-
sequence of several production cell lines now able option for researchers making the switch
available, it is possible to efficiently engineer from transient expression to stable lines.
these genomic sequences with nucleases.
Customized nucleases such as zinc finger
nucleases (ZNFs), transcription activator- TRANSFECTING CELLS
like effector nucleases (TALENs), and The gene encoding the protein of interest
clustered regularly interspaced short palin- must be inserted into a cell before the
dromic repeats (CRISPR)-associated (CAS) protein can be produced. Many transfection
could be an advantageous choice for the future. procedures are available, broadly classified
The CRISPR/Cas system has been success- as chemical, viral, and physical methods
fully used for genome editing in CHO during (Kim & Eberwine, 2010). The approach
cell line development (Lee, Grav, Lewis, & offers high transfection efficiency, low cell
Faustrup Kildegaard, 2015), and recently it toxicity, and reproducibility. The choice of
has been reportedly used for site-specific method depends on the cell type, the amount
integration of a therapeutic gene in CHO (Lee, of protein required, and the cost involved.
Kallehauge, Pedersen, & Kildegaard, 2015). In choosing an approach, the downstream
Transposase enzymes can integrate recom- application must also be considered: it may
binant genes into the genomes of mammalian be possible to use a technique successfully
cell lines. When transposases recognize short at the small scale that is not cost-effective at
inverted DNA sequences, they excise DNA large scale. All transfection approaches have
that is flanked by these repeats and inte- their advantages and drawbacks, and once a
grate it at a new location in the cell genome method is selected, critical parameters need to
(Fig. 2A). Transposases can be used to cre- be optimized at each stage of the development
ate stable cell lines by co-transfecting cells process (Kim & Eberwine, 2010).
with a donor plasmid and some form of the
transposase. The donor plasmid contains the Chemical Methods
gene of interest and a selection marker flanked Chemical transfection methods, in which
by the repeats, called a transposon. The trans- a chemical is added to the DNA to facili-
posase can be transfected as a helper plas- tate entry into the cell, have been the most
Hunter et al.
mid coding for the transposase, or the purified widely used since their introduction in 1973.
10 of 28
Current Protocols in Protein Science
Figure 2 Transposase enzymes allow rapid integration of recombinant genes into genomes of
cultivated mammalian cells. (A). Transposases can excise DNA flanked by short inverted DNA
sequences recognized by the transposase and integrate them at a new location in the cell genome,
keeping the entire region (expression cassette) between the short-inverted DNA sequences intact.
Without the transposase, the expression cassette can be truncated or scrambled, resulting in
transgene-transgene fusion (TG/TG fusion) sequences. This approach has been successfully
used to rapidly generate stable pools with volumetric productivities higher than those produced by
traditional random integration methods. (B). Targeted Locus Amplification (TLA) sequencing data
of transgenes and their integration sites in a stable cell line. The table shows the proportions of
identified integration sites that were accomplished via transposition and via random integration,
as well as the number of TG/TG fusion sequences that result from concatemerization. The data
shows that the number of TG/TG fusions is far greater in the cells without transposase. (C). Rapid
selection of clonal lines with consistent productivity from stable pools. The highest expressing
clones are selected from pools giving high protein productivities of approximately 4 g/L (y-axis).
(D). The timeline for generating stable pools can be as little as 2 to 4 weeks from transfection to
protein recovery, establishing cell pools suitable for protein production and selection of clonal cell
lines for research cell banking (RCB).

CaCl2 (Graham & van der Eb, 1973) or DEAE- ability to transfect a variety of cell types at
dextran (McCutchan & Pagano, 1968; Vaheri high efficiency. A wide range of cationic lipid
& Pagano, 1965) are the two oldest approaches products has become commercially available,
and are very cost-effective but may suffer from such as TransFectinTM (Biorad), FuGENE R

low transfection efficiency. Transfection effi- HD (Promega), FectoPro (Polyplus transfec-


ciency is the percentage of cells transformed tion) and Lipofectamine (ThermoFisher). Al-
and is typically assessed using a report plas- ternatively, polyplexes like Polyethelyimine
mid. DEAE-dextran is not suitable to generate (PEI) are an economical method of transfec-
stable lines, and both approaches cause tox- tion (Rajendra, Kiseljak, Baldi, Hacker, &
icity to the cells (Gluzman, 1981). Cationic Wurm, 2011). Different chemical forms of
lipids are the most popular chemical transfec- PEI are available from a variety of suppli-
tion method and can be scaled up to thou- ers, and many can generate transfection ef-
sands of liters. Their main advantage is their ficiencies of 100% with little cellular toxicity. Hunter et al.

11 of 28
Current Protocols in Protein Science
However, some growth media are not com- transfer into cells and are the most efficient
patible with PEI transfection as they contain vehicle for DNA transfer. However, there are
components that interfere with PEI activity. significant safety issues regarding their use,
This can be mitigated by a media change or and they are not permitted for the develop-
cell wash before transfection, or by select- ment of recombinant proteins that may en-
ing a suitable growth media initially. PEI is ter the clinic. Lentiviral vectors are capable
scalable from 1 ml or less to many thousands of transducing a broad range of cell types and
of liters (Ye et al., 2009). The processes by have been shown to generate stable cell lines in
which each of these approaches introduces both HEK-293 and CHO cells. One significant
DNA into a cell are similar. Nucleic acids drawback to the lentiviral system is that the
are negatively charged due to their polyphos- capsid is limited to a packaging size of approx-
phate backbone, and these positively charged imately 10 kb, restricting the size of the protein
chemicals interact with the DNA forming that can be made (Kumar, Keller, Makalou, &
complexes. Presumably, these positively Sutton, 2001). The BacMam System uses a
charged complexes attach to the negatively baculovirus, a virus that infects insect cells,
charged cell surface (sialic acid residues), trig- that has been modified to swap promoter el-
gering cell encapsulation by endocytosis and ements that are active in insect cells to ones
phagocytosis. The DNA is transported to the that are active in mammalian cells, such as the
nucleus of the cells, where the cells transcrip- cytomegalovirus (CMV) promoter (Fornwald,
tion and translation machinery produces the Lu, Wang, & Ames, 2007). The viruses are
protein of interest (Gutierrez-Granados et al., generated and amplified in insect cells and are
2018). then used to infect mammalian cells, where the
target protein will be produced. Improved Bac-
Mam vectors have been generated, express-
PHYSICAL METHODS ing vesicular stomatitis virus glycoprotein G
Physical transfection methods include elec- in the viral envelope to make the infection of
troporation, direct injection, biolistic particle mammalian cells more efficient. These sys-
delivery, and laser-based transfection. Electro- tems have proved popular as the baculovirus
poration is the most widely used approach for can deliver its payload to a large variety of
delivering DNA to cells for protein production. cell types. However, the amount of virus re-
It uses pulsed electric fields to introduce DNA quired to infect mammalian cells effectively
into the cells (Longo, Kavran, Kim, & Leahy, often needs large baculovirus production runs,
2013). Cells and the plasmid containing the which take a few weeks and have their inher-
gene of interest are suspended in a conduc- ent costs (Barsoum, Brown, McKee, & Boyce,
tive solution enclosed in an electrical circuit. 1997). BacMam is widely used in the mem-
A short electrical pulse is discharged through brane protein structural biology field, possi-
the suspension, disturbing the phospholipid bi- bly because insect cell culture has been heav-
layer of the cell membrane, creating tempo- ily utilized by this group, making it a smooth
rary pores through which the DNA can pass transition between systems (Dukkipati, Park,
(Shigekawa & Dower, 1988). Electroporation Waghray, Fischer, & Garcia, 2008).
is applicable to both stable and transient trans-
fection and transfects many cells in a short pe-
riod once optimal electroporation conditions SELECTION METHODS
have been determined. The primary disadvan- Selection techniques for generating stable
tages are a lack of scalability and high levels cell lines are broadly grouped into drug re-
of cell death caused by the high voltage pulses. sistance, metabolic rescue, or physical meth-
Equipment to electroporate cells are available ods, and these techniques can be used alone
from many vendors, including ThermoFisher, or in combination. A typical cell line develop-
Biorad, and Eppendorf. MaxCyte has devel- ment program starts with a transgenic plasmid
oped a scalable transfection system using Flow containing the gene of interest and a selection
ElectroporationTM . However, this technology marker. After integration into the cell genome,
is effective only up to 200 liters, significantly a selection agent is used to kill off the cell pop-
short of the thousands of liters achieved by the ulation that has little to no expression of the
chemical approach. transgene. In some instances, selection pres-
sure is increased to enable gene amplification
Viral Methods and elevate protein expression. When creating
Viral vector systems consist of a defective clonal cell lines, cells that survive the selec-
Hunter et al.
virus that encapsulates the gene of interest for tion process are diluted to single cells and ex-
12 of 28
Current Protocols in Protein Science
Figure 3 Selection drug kill curves are shown for three commonly used antibiotics (A). Blasticidin,
(B). Puromycin and (C). Zeocin on HEK-293 cells grown for 6 days and viability measurements taken
at day 0, 1, 2, 3 and 6. Percent viability is shown for each time-point along the y-axis. Untreated HEK
293 cells are used as positive controls. In order to generate a stable line expressing a transgene it is
important to generate selection-drug kill curve to determine the minimum amount of drug required to
kill cells that were not transfected or transduced with a drug resistance marker.

panded as clonal populations. Timescales for fication technology (Kaufman & Sharp, 1982).
the generation of a stable cell line, in which Glutamine synthetase knockout cell lines are
the gene of interest becomes integrated into available for CHO (L. Fan et al., 2012) in
the host cell genome, can also vary widely be- which the GS gene is deleted, requiring the cell
tween selection methods. culture media to be supplemented with glu-
Positive selection markers that rely on toxic tamine for the cells to grow. Transformation of
agents were the first approach to be devel- the genes of interest into a cassette with genes
oped. These drug resistance strategies are for the expression of exogenous glutamine
highly efficient, with selection times of only synthetase allows for the rescue of transformed
a few days. HEK-293 and CHO cells are sen- cells in glutamine-deficient media (Knox et al.,
sitive to drugs such as Zeocin, Puromycin, 2013). Selective pressure can be increased
Hygromycin B, Ouabain, and G418 (Lanza, by using the drug methionine sulphoximine
Kim, & Alper, 2013; Treschow et al., 2007). (MSX), which inhibits the GS enzyme, or
Some cell strains, such as HEK-293T, are by weak promoters driving GS production, or
resistant to certain drugs, so optimal antibi- even by destabilizing the GS protein by the ad-
otic selection must be determined for a given dition of PEST sequences, which act as a signal
cell and media combination to generate a for protein degradation (L. Fan et al., 2013).
kill curve. For any drug resistance selection Increasing selective pressure can lead to gene
method, the generation of a selection-drug kill amplification and even higher expression lev-
curve will determine the minimum amount of els of the target protein. These processes force
drug required to kill cells that were not trans- the cell to make more of the protein of inter-
fected or transduced (Fig. 3). The modes of est, but some of these amplification techniques
action of these drugs are entirely different. can lead to genomic instability. Cell line gen-
Zeocin is a bleomycin analog that binds and eration using GS is reported to be faster than
cleaves DNA (Oliva-Trastoy, Defais, & Larmi- the related dihydrofolate reductase selection
nat, 2005). G418 and Hygromycin are amino- system.
glycosides that kill eukaryotic cells by binding The CHO-DG44 line is a dihydrofolate re-
ribosomal components and inhibiting transla- ductase (DHFR) deficient cell line, in which
tion (Bernard, Krammer, & Rowekamp, 1985). selection is based on media that does not con-
Puromycin is an aminonucleoside that also dis- tain thymidine, glycine or hypoxanthine, as
rupts translation (Azzam & Algranati, 1973), DHFR is critical for the synthesis of these
and Ouabain is a clinically used cardiac gly- molecules (Cacciatore, Chasin, & Leonard,
coside and selective Na+ /K+ -ATPase inhibitor 2010). Transformation of cells with the gene
(Treschow et al., 2007). of interest and the gene for the DHFR enzyme
Metabolic selection markers, utilizing cell allows for growth in thymidine, glycine or hy-
strains that are deficient in critical biosynthetic poxanthine deficient media which is then cou-
pathways, are commonly used in high-level pled with methotrexate (MTX) drug selection
expression systems. Cell line development and complementary gene amplification (Vish-
technologies used by most biopharmaceutical wanathan et al., 2014). Other metabolic selec-
companies are usually based off either the glu- tion pathways have been investigated such as
tamine synthetase (GS) system (Bebbington folate and purine synthesis, and given the rel-
et al., 1992) or the methotrexate (MTX) ampli- ative ease of CRISPR/CAS technologies and Hunter et al.

13 of 28
Current Protocols in Protein Science
genome sequencing, it seems likely that many a promoter, a multi-cloning site (MCS),
new selection methods will soon become avail- and a 3’ untranslated region (3’UTR) con-
able (Costa et al., 2012; Rothem, Berman, taining a polyadenylation (polyA) sequence
Stark, Jansen, & Assaraf, 2005). (Kaufman, 2000). Other elements often
Each selection method will result in a pop- include enhancers, introns, viral amplifiers,
ulation of cells producing target proteins at IRES elements, selection markers, origins
different levels. If a stable pool is not suffi- of replication, and chromatin remodeling
cient for production (stable pools can gener- factors (Fig. 4A). Accordingly, different
ate grams per liter of antibodies), then further vector design strategies mix and match these
selection techniques can be used to create a elements to produce large amounts of stable
clonal cell line. Limited dilution is the process messenger RNA and, by extension, proteins.
of making serial dilutions of a cell stock and Enhancers and promoters are upstream of
then dispensing a cell suspension into individ- the MCS and cooperate to determine the
ual wells of multi-well plates to isolate a single rate of transcription. A promoter is an ar-
cell in each well. This cell will proliferate, cre- rangement of short regulatory sequences that
ating a group of cells that are clonally identi- attract sequence-specific transcription factors
cal. Limited dilution is tedious and inefficient that drive transcription initiation (Dalton &
but does not require sophisticated equipment. Barton, 2014). Enhancers are sequences that
Fluorescence-activated cell sorting (FACS) is bind activator proteins that increase the rate
utilized to sort a cell suspension into single of transcription and can be adjacent or far
cells (Misaghi et al., 2016). FACS can be used from the promoter they influence. Commonly
to sort a population of cells, selecting cells used promoters are the CMV, Ef1α, CAG, and
with high target protein production levels by SV40 (Dalton & Barton, 2014). Many viral
linking the gene of interest to a fluorescent promoter regions contain proximal enhancers,
marker, such as in the IRES-GFP system, or such as CMV and SV40, frequently incorpo-
by cell surface display, or by ‘cold’ sorting to rated into vector cassettes that are in some
capture protein secretion in the membrane of cases cell-specific (Brown, Sweeney, Main-
the cell, which can then be probed by a fluo- waring, & James, 2014; Mariati, Ho, Yap, &
rescent secondary antibody (Brezinsky et al., Yang, 2010).
2003). In each of these techniques, the level of Simultaneous expression of the light and
fluorescence associated with a cell correlates heavy chain of a monoclonal antibody can
to the level of production of the target pro- be achieved using two independent plasmids.
tein, so that only highly producing cells will However, this approach is heavily influenced
be captured. Cells may also be sorted in ad- by transfection efficiency, the copy number of
herent culture using lasers to induce cell death each plasmid that enters the cell, and, in the
of non-producers (Koller et al., 2004) . Fluo- case of stable cell lines, genomic integration
rescent selection is also utilized in semi-solid sites. An alternative approach is to use a single
media, where target proteins being secreted vector with two independent promoters. How-
from the cell remain in close proximity (Caron ever, transcription interference can occur, in
et al., 2009; Mangalampalli et al., 2015). Prob- which the influence of a strong transcriptional
ing with a fluorescent antibody against the se- process reduces the expression of the sensitive
creted protein reveals high-producing clones promoter. Transcriptional interference can be
that are automatically picked and transferred solved with the use of an internal ribosome
into multi-well plates for further growth (Dhar- entry sites (IRES), either of cellular or viral
shanan, Chong, Swee Hung, Zamrod, & Ka- origin. The IRES approach involves the design
mal, 2011). of a bicistronic construct where the two genes
are separated by an IRES sequence, allowing
two different proteins to be translated from
EXPRESSION VECTORS a single mRNA. This intervening IRES
Expression vectors, also known as expres- sequence functions as a ribosome-binding site
sion constructs, are critical to the production for efficient cap-independent internal initia-
of proteins, as they are the vehicle by which tion of translation. The IRES system offers the
the gene that codes for the protein of interest advantage of using a single promoter for the
is carried into the target cell. These vectors expression of both polypeptides of the mon-
must contain the essential elements by which oclonal antibody, and it allows the expression
the cells’ machinery recognizes the DNA and of the gene of interest in the presence of a
begins transcription and translation of the selection marker for stable line generation
Hunter et al.
protein of interest. These elements include (Li et al., 2007).
14 of 28
Current Protocols in Protein Science
Figure 4 (A). Expression vectors, or expression constructs, are critical to the production of
proteins. These vectors must contain essential elements, including a promoter, multiple cloning
site (MCS), and polyadenylation sequence by which the cells’ machinery recognizes the DNA and
begins transcription and translation of the protein of interest. They may also include additional
elements such as enhancers, introns, viral amplifiers, IRES elements, selection markers, origins
of replication and chromatin remodeling factors, which can increase expression levels. Modular
vector designs utilizing common cloning sites allow different vector design strategies to mix and
match these elements to optimize protein expression in the cell line of choice. (B). Expression of
trastuzumab in different vector configurations is shown. Vector configurations tested are shown
along the x-axis and protein productivity in mg/L is shown along the y-axis. The features of each
vector are listed in Table 1. Selection of a vector is complex: no single expression vector works
for all systems, therefore a systematic approach that optimizes each element in combination with
others needs to be undertaken. Design of Experiments (DOE) was used by ATUM to design a
set of vectors containing elements like those in Figure 4A to be highly productive in a particular
mammalian host compared to standard off the shelf vectors.

Just as the promoter initiates transcription, Proudfoot, 1987; Goodwin & Rottman, 1992;
the 3’ untranslated region is imperative for Lanoix & Acheson, 1988; Schek, Cooke, &
transcript termination. The 3’ end of the RNA Alwine, 1992). Termination and polyadeny-
terminates with a polyA sequence that pro- lation of an mRNA are a coordinated process.
motes nuclear export, stability, and efficient The terminator, a sequence-based element, de-
translation (Edmonds, 2002; Wickens, Ander- fines the end of the transcript, creating a free
son, & Jackson, 1997). Commonly used mam- 3’ end and initiates release of the mRNA from
malian terminators are the SV40, hGH, BGH the transcriptional machinery. The free 3’ end
and rbGlob polyA sequences that provide is then available for the addition of the polyA
both polyadenylation and termination (Gil & tail. Hunter et al.

15 of 28
Current Protocols in Protein Science
The addition of an intron or an untranslated ‘weaker’ promoter, or by engineering cells to
exon sequence upstream from the initiation have enhanced properties (B. Estes et al., 2015;
codon of the gene of interest has been shown Rajendra et al., 2012).
to induce higher levels of protein expression Inducible vectors are used in the produc-
than plasmids without these elements (Buch- tion of target proteins that may be toxic to cell
man & Berg, 1988; Lacy-Hulbert et al., 2001; growth, such as integral membrane proteins.
Melcher, Grosch, & Hasilik, 2002). These These systems allow transfected cells to grow
untranslated exons contain splicing elements uninduced at a similar rate to the parental cell
that are thought to improve mRNA transport line, and then, upon induction, synthesize the
from the nucleus to the cytoplasm, or enhance desired protein. Inducible transient expression
RNA stability and or half-life, thus provid- systems typically rely on the co-transfection of
ing a greater abundance of the transcript. As a plasmid that expresses a regulatory protein
a result, introns between the promoter and which binds to operator sequences upstream
MCS of commercially available mammalian of the gene of interest. Alternatively, stable
expression vectors are often included. For ex- cells lines can be developed that constitutively
ample, Intron A in combination with hCMV express the regulatory protein. Commercially
IE1 enhancer-promoter can radically increase available inducible systems include the Tet
levels of translatable mRNA, leading to higher operon (from Clontech) which can be used
cellular productivity. to turn on expression in the presence of the
The effects of these additional elements in small molecule drug doxycycline (Dox; Do-
expression vectors depend on whether the gene rai et al., 2012; Gaillet et al., 2010), although
of interest is integrated into the genome or ex- there are many more systems available. The
pressed transiently. Human CMV promoters Cumate inducible system can be used sim-
may be more active than the mouse promot- ilarly (Baranyi et al., 2013). Other systems
ers in stable cell lines, as they are less prone have utilized temperature, isopropyl β-D-1-
to epigenetic silencing. Elements such as uni- thiogalactopyranoside (IPTG), metals, light,
versal chromatin opening elements (UCOE) and insect hormones such as Ecdysone (Ah-
may inhibit chromatin condensation around madi et al., 2016; Casales et al., 2010; Kwaks
target integrated genes, inhibiting transcrip- & Otte, 2006; Neville et al., 2017; Saunders
tional silencing (Betts, Croxford, & Dickson, et al., 2015).
2015; Rocha-Pizana, Ascencio-Favela, Soto- No single expression plasmid works for all
Garcia, Martinez-Fierro, & Alvarez, 2017). systems and selecting one will be dependent
Woodchuck hepatitis post-transcriptional reg- upon the cell line, the type of expression, and
ulatory element (WPRE) and Hepatitis B virus perhaps other factors not considered in this
regulatory element (HPRE) may enhance post- review. Traditional protein expression vectors
transcriptional processing or export of mRNA employed by many are an assembly of nat-
leading to increased protein production (Klein urally occurring parts chosen from a limited
et al., 2006). pool of available enhancers, promoters, termi-
It should be noted that all of these high pro- nators, translation initiation signals, selection
ductivity expression vectors are not without markers, etc. Many of these vectors are merely
their drawbacks (Halff, Versteeg, Brondijk, & functional constructions used either because of
Huizinga, 2014). The quality of protein pro- historical precedent or because an academic
duced must be assessed as well as the quan- lab or company have identified a new regula-
tity. Decreasing effective promotor strength tory element. To know which combination of
has been demonstrated to lead to reduced pro- features will provide the best productivity for
tein aggregation. Intron sequences may have the gene of interest in the chosen expression
cryptic donor or acceptor sites in the target host, a panel of vectors needs to be tested in
gene leading to mRNA splice variants that do parallel to empirically determine the best solu-
not code for the target protein. High rates of tion, which can be accomplished using suites
production may swamp the post-translational of vectors with common cloning sites. While
modification apparatus leading to heteroge- the number of potentially useful elements con-
neous product formation or in cellulo aggre- tinues to expand, a systematic approach needs
gation, like Russell bodies (Hasegawa, Woods, to be undertaken to optimize each element
Kinderman, He, & Lim, 2014). Some of these in combination with the others to maximize
problems may be ameliorated by codon op- vector properties. Optimization tools such as
timization, by stuffer DNA, by choosing a DOE can be used to provide a framework for
Hunter et al.

16 of 28
Current Protocols in Protein Science
Table 1 Features for Each Vector Displayed in Figure 4B

Viral Viral
Origin of replication
Vector ID Enhancer Promoter Intron WPRE replication protein
pTT5 CMV CMV synthetic N/A OriP N/A
pCDNA3.4 CMV CMV N/A yes N/A N/A
1 CMV CMV CMVa N/A OriP EBNA
2 CMV CMV CMVa N/A OriP EBNA
3 CMV CMV CMVa N/A OriP EBNA
4 CMV CMV CMVa N/A OriP EBNA
5 CMV CMV CMVc N/A N/A N/A
6 CMV CMV CMVa N/A SV40 SV40 T
antigen
7 CMV CMV CMVa N/A N/A N/A
8 CMV CMV CMVa N/A OriP N/A
9 CMV CMV CMVa N/A OriP EBNA
10 CMV CMV synthetic N/A N/A N/A
11 synthetic synthetic synthetic N/A N/A N/A
12 synthetic synthetic synthetic N/A N/A N/A
13 N/A synthetic synthetic N/A OriP EBNA
14 synthetic synthetic N/A N/A OriP EBNA
15 synthetic CMV N/A N/A N/A N/A
16 synthetic CMV synthetic N/A N/A N/A
17 synthetic CMV synthetic N/A N/A N/A
18 N/A synthetic synthetic N/A N/A N/A
19 synthetic synthetic N/A N/A N/A N/A
20 CMV CMV N/A N/A N/A N/A
21 synthetic CMV CMVc N/A N/A N/A
22 CMV CMV CMVa N/A OriP EBNA
23 SV40 SV40 N/A N/A N/A N/A

CMV stands for cytomegalovirus, as the promoter and enhancer are originally from the genome of the human cy-
tomegalovirus. The SV40 promoter and enhancer were taken from the genome of simian virus 40. The synthetic
enhancers, promoters, and introns are of rodent origin. The CMVa and CMVc introns are two different regions from the
CMV enhancer/promoter that have been shown to increase expression in mammalian cells. WPRE stands for Woodchuck
Hepatitis virus posttranscriptional regulatory element, which has been shown to increase protein yield when present
in the 3’ untranslated region. Viral origins of replication and their viral replication proteins allow episomal replication
of plasmids. The OriP/EBNA pair is from the Epstein-Barr virus, and the SV40/SV40 T-antigen pair is from Simian
virus 40.

experimentation in which several element GENE OF INTEREST


combinations can be simultaneously tested. In OPTIMIZATION
conjunction with de novo DNA synthesis, it is Considerable effort has been made to
possible to rapidly screen independently de- improve the expression of the gene of in-
signed constructs based on their performance terest (GOI), including the development of
(Fig. 4B, Table 1). The outcome of these ex- mammalian expression hosts, cell culture
periments allows the researcher to purchase, media improvements, construction of con-
with confidence, a vector that will be highly trolled bioreactors, development of rapid and
productive in a defined mammalian host. extended expression methods, and vector
Hunter et al.

17 of 28
Current Protocols in Protein Science
optimization. Despite these advances, one shull, & Govindarajan, 2006). Furthermore,
underlying subject remains to be discussed, they provide the researcher with the oppor-
the GOI itself. The open reading frame used tunity to manipulate their sequence to include
to express the protein of interest goes far or remove sequence elements such as restric-
beyond merely stating the order of amino tion sites. A well-designed optimization of
acids. In its native context, it may contain sec- a gene is a big challenge; however, a large
ondary structures, mRNA that might inhibit variety of software is available in the pub-
ribosome processivity, alternative splicing lic domain or as commercial sequence pack-
sites, sequence elements signaling mRNA ages. The software for optimization must take
degradation, or codons that are rarely used in into consideration many parameters spanning
an expression host. Therefore, the variables the entire gene of interest, including short se-
that affect the open reading frame of the GOI quence motifs, splice site recognition patterns,
and its ability to produce large amounts of GC-content and codon usage (Villalobos et al.,
protein in mammalian cells can be numerous 2006).
(Gustafsson et al., 2012; Welch, Villalobos, In addition to the optimization of the gene
Gustafsson, & Minshull, 2009). De novo DNA that will produce the recombinant protein of
synthesis provides the researcher with the interest, we must consider where the protein
ability to create a full-length gene considering will be present once made. The widespread
these parameters and controlling for the use of mammalian cells has been dominated
presence or absence of specific restriction by the production of secreted proteins that re-
sites or motifs. quire a signal sequence that directs the pro-
De novo gene synthesis allows fast and ef- tein of interest outside of the cell. These are
ficient construction of genes tailored for a spe- typically short 20-30 hydrophobic amino acid
cific application, as well as the design of an helices which are recognized by cellular ma-
“optimal” gene. One interpretation of “opti- chinery and used to target the protein of in-
mal” could mean designing a DNA sequence terest down the correct pathway. Signal se-
that increases the cell’s ability to express a quences may also be present on proteins that
recombinant protein at high levels by opti- are not secreted from the cell but embedded
mizing codon choices. Many examples ex- into the cell membrane, such as integral mem-
ist demonstrating the impact of codon usage brane proteins or cell surface receptors. It has
on gene expression. Specifically, research has been reported that the expression of a recom-
demonstrated that gene expression levels cor- binant protein does not always correlate with
relate with the use of optimal codons, codons its mRNA levels (L. M. Barnes, Bentley, &
that have the highest tRNA gene copy num- Dickson, 2004) and a rate-limiting step can
ber. They demonstrate that there is a high fre- be the secretory pathway. Intuitively, the best
quency of optimal codons in highly expressed choice of a signal sequence may be the pro-
genes, so a simple strategy for increasing ex- teins’ native signal. However, testing a panel
pression of the protein of interest would be of commonly utilized signal sequences is de-
to construct the GOI with optimal codons sirable (Fig. 5). Not surprisingly, the signal se-
(Lavner & Kotlar, 2005). Another optimiza- quence can have a dramatic effect on protein
tion strategy would be to utilize codon usage productivity, with fourfold enhancement of ex-
bias and assign the most highly used codons pression levels being reported (Kober, Zehe, &
to all instances of the amino acid sequence. Bode, 2013; Stern, L.C., Trobe, Ravneberg, &
This line of reasoning means that an ideal Pryme, 2007).
codon is defined by its frequency, which is Recombinant proteins are commonly ex-
often measured by the Codon Adaption Index pressed in eukaryotic expression systems to
(CAI) score (Sharp & Li, 1987). Such an ap- ensure the formation of disulfide bridges and
proach has many drawbacks, including gener- proper glycosylation. Antibody and antibody-
ating a high codon concentration subset of the like molecules are examples of proteins that
tRNA population, resulting in an imbalanced are relatively easily made in mammalian cells
tRNA pool, skewing codon usage, and in- and are easily purified by Protein A or Protein
creasing translational error (Plotkin & Kudla, G resins. However, some recombinant pro-
2011). This approach has been superseded by teins, sub-domains, and mutant protein ver-
the development of advanced algorithms that sions can be plagued with issues regarding
not only take these issues into account, but misfolding, aggregation, or lack of expression
can also avoid repetitive elements and mRNA entirely. Fusion tags are frequently used to ad-
structure (Villalobos, Ness, Gustafsson, Min- dress these problems and provide a convenient
Hunter et al.

18 of 28
Current Protocols in Protein Science
Figure 5 Effect of different secretion signals on the expression of antibody light chain (LC)
and heavy chain (HC) is shown. Secretion signals tested are shown on the x-axis and model
regression weights shown on the y-axis. A total of 23 different secretion signals were tested on
the light and heavy chains of 5 different antibodies. ATUM utilizes DOE and machine learning
algorithms that assign model regression weights based off the mean protein titer of each antibody
(y-axis) to different secretion signals (x-axis), predicted for their effect on (A). light (LC) and (B).
heavy chains (HC) of an antibody. The model regression weights of each signal sequence are
represented as a cluster of the 5 antibodies on which it has been tested. Positive contributions
have positive regression weights, and the magnitude of the weight corresponds to the magnitude
of the effect of the secretion sequence. As the data shows, some secretion signals can have
positive effects on one antibody and negative effects on another, while others have net positive
effects on all 5 antibodies tested.

means for single step purification. This review known. But beyond deciding on the protein of
will not extensively cover these tags. Com- interest, it can be difficult to choose a starting
monly used fusion partners include the con- point for process development.
stant domain of IgG (the Fc region), maltose The numerous variables that are involved
binding protein (MBP), small ubiquitin-like in mammalian protein expression are interde-
modifier (SUMO), and human serum albumin pendent, so changing one can have unexpected
(HSA) tag (Bokhove et al., 2016; Carter et al., downstream effects. A common starting point
2010). All have been shown to improve yield, is selection of the expression host, which has
solubility, or both for the protein that they were a direct connection to the culture media, trans-
fused to, but the extent of their influence does fection method, and expression vector used.
appear to be protein specific. The Fc tag does HEK-293 cells are often used for transient ex-
cause artificial dimerization of fused proteins, pression due to its ease of transfectability and
which may induce aggregation with certain high titers. CHO cells are more commonly
partners. As many of these tags are large, it used for stable cell line expression, as most
is often necessary to remove them after the fu- FDA-approved biotherapeutics are produced
sion protein has been purified. The most com- in CHO. It is not unusual for a biotherapeutic
mon solution is to engineer a protease cleavage development process to start with HEK-293
site between the solubility tag and the partner expression and move into CHO once screening
recombinant protein. has been done on several protein versions. This
switch requires further development of the me-
dia, expression vector, and transfection and se-
CONCLUSIONS lection methods. Scaling up the culture size as
With the continuing growth in demand for development progresses affects protein char-
recombinant proteins, researchers are seeking acteristics like glycosylation, and these must
rapid methods to produce their protein of in- be accounted for and optimized as well.
terest. To meet these needs there are several No choice in the development of a mam-
mammalian expression platforms that can be malian expression system is made in isolation,
bought off the shelf from commercial suppli- as they tend to have both upstream and down-
ers. However, as most of the information re- stream effects (Fig. 1). Despite the increasing
garding the composition of commercial kits is number of approaches for expressing the gene
proprietary, these platforms do not lend them- of interest in a mammalian platform, there is
selves to easy troubleshooting. In order to no system capable of meeting all challenges.
fully understand the outcome of a mammalian The scale and expectations for those requiring Hunter et al.
expression system, all of the inputs must be
19 of 28
Current Protocols in Protein Science
material for biological research greatly differ Barnes, D., & Sato, G. (1979). Growth of a hu-
from the requirements for those needing ma- man mammary tumour cell line in a serum-
free medium. Nature, 281(5730), 388–389. doi:
terial for drug discovery or development. De-
10.1038/281388a0.
veloping a system that results in the protein of
Barnes, L. M., Bentley, C. M., & Dickson, A.
interest at the correct scale, with correct char-
J. (2004). Molecular definition of predictive
acteristics and biologic activities, at the right indicators of stable protein expression in re-
price, can take months, or even years and is combinant NS0 myeloma cells. Biotechnol-
outside the scope of this review. This process ogy and Bioengineering, 85(2), 115–121. doi:
can be accelerated with the help of a contract 10.1002/bit.10893.
research organization (CRO) or contract man- Barsoum, J., Brown, R., McKee, M., & Boyce,
ufacturing organization (CMO). F. M. (1997). Efficient transduction of mam-
malian cells by a recombinant baculovirus hav-
ing the vesicular stomatitis virus G glycoprotein.
ACKNOWLEDGEMENTS Human Gene Therapy, 8(17), 2011–2018. doi:
The authors would like to thank Miles 10.1089/hum.1997.8.17-2011.
Scotcher, Ferenc Boldog, Alan Villalobos, and
Bebbington, C. R., Renner, G., Thomson, S., King,
Medini Gore for proof reading and intellectual D., Abrams, D., & Yarranton, G. T. (1992).
input. High-level expression of a recombinant anti-
body from myeloma cells using a glutamine syn-
LITERATURE CITED thetase gene as an amplifiable selectable marker.
Ahmadi, M., Damavandi, N., Akbari Eidgahi, M. Biotechnology (N Y), 10(2), 169–175.
R., & Davami, F. (2016). Utilization of site-
Bernard, H. U., Krammer, G., & Rowekamp,
specific recombination in biopharmaceutical
W. G. (1985). Construction of a fusion gene
production. Iranian Biomedical Journal, 20(2),
that confers resistance against hygromycin
68–76.
B to mammalian cells in culture. Experi-
Asher, D. M. (1999). Bovine sera used in the man- mental Cell Research, 158(1), 237–243. doi:
ufacture of biologicals: Current concerns and 10.1016/0014-4827(85)90446-X.
policies of the U.S. Food and Drug Adminis-
Betts, Z., Croxford, A. S., & Dickson, A. J.
tration regarding the transmissible spongiform
(2015). Evaluating the interaction between
encephalopathies. Developments in Biological
UCOE and DHFR-linked amplification and
Standardization, 99, 41–44.
stability of recombinant protein expression.
Azzam, M. E., & Algranati, I. D. (1973). Mech- Biotechnology Progress, 31(4), 1014–1025. doi:
anism of puromycin action: Fate of ribo- 10.1002/btpr.2083.
somes after release of nascent protein chains
Blasey, H. D., Aubry, J. P., Mazzei, G. J., &
from polysomes. Proceedings of the Na-
Bernard, A. R. (1995). Large scale transient ex-
tional Academy of Sciences of the United
pression with COS cells. Cytotechnology, 18(3),
States of America, 70(12), 3866–3869. doi:
183–192. doi: 10.1007/BF00767766.
10.1073/pnas.70.12.3866.
Bokhove, M., Sadat Al Hosseini, H., Saito, T., Dio-
Backliwal, G., Hildinger, M., Kuettel, I., Dele-
guardi, E., Gegenschatz-Schmid, K., Nishimura,
grange, F., Hacker, D. L., & Wurm, F. M.
K., . . . Jovine, L. (2016). Easy mammalian
(2008). Valproic acid: A viable alternative to
expression and crystallography of maltose-
sodium butyrate for enhancing protein expres-
binding protein-fused human proteins. Jour-
sion in mammalian cell cultures. Biotechnol-
nal of Structural Biology, 194(1), 1–7. doi:
ogy and Bioengineering, 101(1), 182–189. doi:
10.1016/j.jsb.2016.01.016.
10.1002/bit.21882.
Bos, A. B., Luan, P., Duque, J. N., Reilly, D.,
Balasubramanian, S., Matasci, M., Kadlecova, Z.,
Harms, P. D., & Wong, A. W. (2015). Opti-
Baldi, L., Hacker, D. L., & Wurm, F. M. (2015).
mization and automation of an end-to-end high
Rapid recombinant protein production from pig-
throughput microscale transient protein pro-
gyBac transposon-mediated stable CHO cell
duction process. Biotechnology and Bioengi-
pools. Journal of Biotechnology, 200, 61–69.
neering, 112(9), 1832–1842. doi: 10.1002/bit.25
doi: 10.1016/j.jbiotec.2015.03.001.
601.
Balasubramanian, S., Rajendra, Y., Baldi, L.,
Brezinsky, S. C. G., Chiang, G. G., Szilvasi, A.,
Hacker, D. L., & Wurm, F. M. (2016). Compar-
Mohan, S., Shapiro, R. I., MacLean, A., . . .
ison of three transposons for the generation of
Thill, G. (2003). A simple method for enriching
highly productive recombinant CHO cell pools
populations of transfected CHO cells for cells
and cell lines. Biotechnology and Bioengineer-
of higher specific productivity. Journal of Im-
ing, 113(6), 1234–1243. doi: 10.1002/bit.25888.
munological Methods, 277(1–2), 141–155. doi:
Baranyi, L., Doering, C. B., Denning, G., Gaut- 10.1016/s0022-1759(03)00108-x.
ney, R. E., Harris, K. T., Spencer, H. T., . . .
Brindley, D., Moorthy, K., Lee, J. H., Mason, C.,
Dropulic, B. (2013). Rapid generation of sta-
Kim, H. W., & Wall, I. (2011). Bioprocess
ble cell lines expressing high levels of ery-
forces and their impact on cell behavior: Im-
thropoietin, factor VIII, and an antihuman
plications for bone regeneration therapy. Jour-
CD20 antibody using lentiviral vectors. Human
nal of Tissue Engineering, 2011, 620247. doi:
Hunter et al. Gene Therapy Methods, 24(4), 214–227. doi:
10.4061/2011/620247.
10.1089/hgtb.2013.002.
20 of 28
Current Protocols in Protein Science
Brown, A. J., Sweeney, B., Mainwaring, D. O., vector. N Biotechnology, 27(2), 138–148. doi:
& James, D. C. (2014). Synthetic promot- 10.1016/j.nbt.2010.02.005.
ers for CHO cell engineering. Biotechnology Cervera, L., Fuenmayor, J., Gonzalez-Dominguez,
and Bioengineering, 111(8), 1638–1647. doi: I., Gutierrez-Granados, S., Segura, M. M., &
10.1002/bit.25227. Godia, F. (2015). Selection and optimization
Buchman, A. R., & Berg, P. (1988). Com- of transfection enhancer additives for increased
parison of intron-dependent and intron- virus-like particle production in HEK293 sus-
independent gene expression. Molecular and pension cell cultures. Applied Microbiology
Cellular Biology, 8(10), 4395–4405. doi: and Biotechnology, 99(23), 9935–9949. doi:
10.1128/MCB.8.10.4395. 10.1007/s00253-015-6842-4.
Butler, M., Imamura, T., Thomas, J., & Thilly, W. G. Chan, S.-Y., & Harris, K. (1997). USA Patent No.:
(1983). High yields from microcarrier cultures U. S. Patent.
by medium perfusion. Journal of Cell Science, Chartrain, M., & Chu, L. (2008). Development
61, 351–363. and production of commercial therapeutic mon-
Butler, M., & Spearman, M. (2014). The choice oclonal antibodies in Mammalian cell expres-
of mammalian cell host and possibilities sion systems: An overview of the current up-
for glycosylation engineering. Current Opin- stream technologies. Current Pharmaceutical
ion in Biotechnology, 30, 107–112. doi: Biotechnology, 9(6), 447–467. doi: 10.2174/13
10.1016/j.copbio.2014.06.010. 8920108786786367.
Cacciatore, J. J., Chasin, L. A., & Leonard, Chaturvedi, K., Sun, S. Y., O’Brien, T., Liu, Y. J., &
E. F. (2010). Gene amplification and vec- Brooks, J. W. (2014). Comparison of the behav-
tor engineering to achieve rapid and high- ior of CHO cells during cultivation in 24-square
level therapeutic protein production using deep well microplates and conventional shake
the Dhfr-based CHO cell selection system. flask systems. Biotechnology Reports, 1-2, 22–
Biotechnology Advances, 28(6), 673–681. doi: 26. doi: 10.1016/j.btre.2014.04.001.
10.1016/j.biotechadv.2010.04.003. Clincke, M. F., Molleryd, C., Zhang, Y., Lindskog,
Cacciatore, J. J., Leonard, E. F., & Chasin, E., Walsh, K., & Chotteau, V. (2013). Very high
L. A. (2012). The isolation of CHO cells density of CHO cells in perfusion by ATF or TFF
with a site conferring a high and repro- in WAVE bioreactor. Part I. Effect of the cell
ducible transgene amplification rate. Jour- density on the process. Biotechnology Progress,
nal of Biotechnology, 164(2), 346–353. doi: 29(3), 754–767. doi: 10.1002/btpr.1704.
10.1016/j.jbiotec.2013.01.016. Cong, L., Ran, F. A., Cox, D., Lin, S., Barretto,
Cachianes, G., Ho, C., Weber, R. F., Williams, S. R., Habib, N., . . . Zhang, F. (2013). Multi-
R., Goeddel, D. V., & Leung, D. W. (1993). plex genome engineering using CRISPR/Cas
Epstein-Barr virus-derived vectors for transient systems. Science, 339(6121), 819–823. doi:
and stable expression of recombinant proteins. 10.1126/science.1231143.
Biotechniques, 15(2), 255–259. Costa, A. R., Rodrigues, M. E., Henriques, M.,
Cain, K., Peters, S., Hailu, H., Sweeney, B., Melton, D., Cunnah, P., Oliveira, R., & Az-
Stephens, P., Heads, J., . . . Dickson, A. eredo, J. (2012). Evaluation of the OSCAR
(2013). A CHO cell line engineered to system for the production of monoclonal anti-
express XBP1 and ERO1-Lalpha has in- bodies by CHO-K1 cells. International Jour-
creased levels of transient protein expression. nal of Pharmaceutics, 430(1-2), 42–46. doi:
Biotechnology Progress, 29(3), 697–706. doi: 10.1016/j.ijpharm.2012.03.028.
10.1002/btpr.1693. Dalton, A. C., & Barton, W. A. (2014). Over-
Caron, A. W., Nicolas, C., Gaillet, B., Ba, I., Pinard, expression of secreted proteins from mam-
M., Garnier, A., . . . Gilbert, R. (2009). Fluores- malian cell lines. Protein Science, 23(5), 517–
cent labeling in semi-solid medium for selection 525. doi: 10.1002/pro.2439.
of mammalian cells secreting high-levels of re- Daramola, O., Stevenson, J., Dean, G., Hatton,
combinant proteins. BMC Biotechnol, 9, 42. doi: D., Pettman, G., Holmes, W., & Field, R.
10.1186/1472-6750-9-42. (2014). A high-yielding CHO transient sys-
Carroll, D. (2011). Genome engineering with zinc- tem: Coexpression of genes encoding EBNA-1
finger nucleases. Genetics, 188(4), 773–782. and GS enhances transient protein expression.
doi: 10.1534/genetics.111.131433. Biotechnology Progress, 30(1), 132–141. doi:
Carter, J., Zhang, J., Dang, T. L., Hasegawa, H., 10.1002/btpr.1809.
Cheng, J. D., Gianan, I., . . . Mehlin, C. (2010). Davies, A., Greene, A., Lullau, E., & Abbott, W. M.
Fusion partners can increase the expression of (2005). Optimisation and evaluation of a high-
recombinant interleukins via transient transfec- throughput mammalian protein expression sys-
tion in 2936E cells. Protein Science, 19(2), 357– tem. Protein Expression and Purification, 42(1),
362. doi: 10.1002/pro.307. 111–121. doi: 10.1016/j.pep.2005.03.012.
Casales, E., Aranda, A., Quetglas, J. I., Ruiz- De Jesus, M., & Wurm, F. M. (2011). Man-
Guillen, M., Rodriguez-Madoz, J. R., Prieto, ufacturing recombinant proteins in kg-ton
J., & Smerdou, C. (2010). A novel system quantities using animal cells in bioreac-
for the production of high levels of func- tors. European Journal of Pharmaceutics
tional human therapeutic proteins in stable and Biopharmaceutics, 78(2), 184–188. doi:
Hunter et al.
cells with a Semliki Forest virus noncytopathic 10.1016/j.ejpb.2011.01.005.
21 of 28
Current Protocols in Protein Science
Dell, B. D., Mcgrew, J. T., Trentalange, M. T., & gineering/Biotechnology, 139, 11–33. doi:
Van, N. K. P. (2004). Methods for increasing 10.1007/10_2013_260.
polypeptide production. In: Google Patents. Fan, L., Kadura, I., Krebs, L. E., Hatfield, C. C.,
Derouazi, M., Girard, P., Van Tilborgh, F., Igle- Shaw, M. M., & Frye, C. C. (2012). Improv-
sias, K., Muller, N., Bertschinger, M., & Wurm, ing the efficiency of CHO cell line genera-
F. M. (2004). Serum-free large-scale tran- tion using glutamine synthetase gene knock-
sient transfection of CHO cells. Biotechnol- out cells. Biotechnology and Bioengineering,
ogy and Bioengineering, 87(4), 537–545. doi: 109(4), 1007–1015. doi: 10.1002/bit.24365.
10.1002/bit.20161. Fan, L., Kadura, I., Krebs, L. E., Larson, J. L.,
Dharshanan, S., Chong, H., Swee Hung, C., Zam- Bowden, D. M., & Frye, C. C. (2013). Develop-
rod, Z., & Kamal, N. (2011). Rapid auto- ment of a highly-efficient CHO cell line gener-
mated selection of mammalian cell line se- ation system with engineered SV40E promoter.
creting high level of humanized monoclonal Journal of Biotechnology, 168(4), 652–658. doi:
antibody using Clone Pix FL system and 10.1016/j.jbiotec.2013.08.021.
the correlation between exterior median in- Fan, Y., Jimenez Del Val, I., Muller, C., Wagt-
tensity and antibody productivity. Electronic berg Sen, J., Rasmussen, S. K., Kontoravdi, C.,
Journal of Biotechnology, 14(2)1–10. doi: . . . Andersen, M. R. (2015). Amino acid and
10.2225/vol14-issue2-fulltext-7. glucose metabolism in fed-batch CHO cell cul-
Dorai, H., Corisdeo, S., Ellis, D., Kinney, C., ture affects antibody production and glycosyla-
Chomo, M., Hawley-Nelson, P., . . . Gan- tion. Biotechnology and Bioengineering, 112(3),
guly, S. (2012). Early prediction of instability 521–535. doi: 10.1002/bit.25450.
of Chinese hamster ovary cell lines express- Fischer, S., Charara, N., Gerber, A., Wolfel, J.,
ing recombinant antibodies and antibody-fusion Schiedner, G., Voedisch, B., & Geisse, S. (2012).
proteins. Biotechnology and Bioengineering, Transient recombinant protein expression in
109(4), 1016–1030. doi: 10.1002/bit.24367. a human amniocyte cell line: The CAP-T(R)
DuBridge, R. B., Tang, P., Hsia, H. C., Leong, cell system. Biotechnology and Bioengineering,
P. M., Miller, J. H., & Calos, M. P. (1987). 109(9), 2250–2261. doi: 10.1002/bit.24514.
Analysis of mutation in human cells by using Fletcher, T. (2005). Designing culture media for
an Epstein-Barr virus shuttle system. Molecu- recombinant protein production. Bioprocess In-
lar and Cellular Biology, 7(1), 379–387. doi: ternational, 3
10.1128/MCB.7.1.379.
Fornwald, J. A., Lu, Q., Wang, D., & Ames, R.
Dukkipati, A., Park, H. H., Waghray, D., Fischer, S. (2007). Gene expression in mammalian cells
S., & Garcia, K. C. (2008). BacMam system using BacMam, a modified baculovirus system.
for high-level expression of recombinant soluble Methods in Molecular Biology, 388, 95–114.
and membrane glycoproteins for structural stud- doi: 10.1007/978-1-59745-457-5_5.
ies. Protein Expression and Purification, 62(2),
160–170. doi: 10.1016/j.pep.2008.08.004. Gaillet, B., Gilbert, R., Broussau, S., Pilotte, A.,
Malenfant, F., Mullick, A., . . . Massie, B.
Eagle, H. (1959). Amino acid metabolism in mam- (2010). High-level recombinant protein pro-
malian cell cultures. Science, 130(3373), 432– duction in CHO cells using lentiviral vec-
437. doi: 10.1126/science.130.3373.432. tors and the cumate gene-switch. Biotechnol-
Eberhardy, S. R., Radzniak, L., & Liu, Z. (2009). ogy and Bioengineering, 106(2), 203–215. doi:
Iron (III) citrate inhibits polyethylenimine- 10.1002/bit.22698.
mediated transient transfection of Chi- Galbraith, D. J., Tait, A. S., Racher, A. J., Birch, J.
nese hamster ovary cells in serum-free R., & James, D. C. (2006). Control of culture
medium. Cytotechnology, 60(1-3), 1. doi: environment for improved polyethylenimine-
10.1007/s10616-009-9198-8. mediated transient production of recombi-
Edmonds, M. (2002). A history of poly A se- nant monoclonal antibodies by CHO cells.
quences: From formation to factors to func- Biotechnology Progress, 22(3), 753–762. doi:
tion. Progress in Nucleic Acid Research 10.1021/bp050339v.
and Molecular Biology, 71, 285–389. doi: Gil, A., & Proudfoot, N. J. (1987). Position-
10.1016/S0079-6603(02)71046-5. dependent sequence elements downstream of
Eibl, R., Loffelholz, C., & Eibl, D. (2014). AAUAAA are required for efficient rabbit beta-
Disposable bioreactors for inoculum pro- globin mRNA 3 end formation. Cell, 49(3),
duction and protein expression. Methods 399–406. doi: 10.1016/0092-8674(87)90292-3.
in Molecular Biology, 1104, 265–284. doi: Gluzman, Y. (1981). SV40-transformed simian
10.1007/978-1-62703-733-4_18. cells support the replication of early
Estes, B., Hsu, Y. R., Tam, L. T., Sheng, J., Stevens, SV40 mutants. Cell, 23(1), 175–182. doi:
J., & Haldankar, R. (2015). Uncovering meth- 10.1016/0092-8674(81)90282-8.
ods for the prevention of protein aggregation and Gomez, N., Ambhaikar, M., Zhang, L., Huang,
improvement of product quality in a transient ex- C. J., Barkhordarian, H., Lull, J., & Gutier-
pression system. Biotechnology Progress, 31(1), rez, C. (2017). Analysis of Tubespins as
258–267. doi: 10.1002/btpr.2021. a suitable scale-down model of bioreac-
Estes, S., & Melville, M. (2014). Mam- tors for high cell density CHO cell culture.
malian cell line developments in speed Biotechnology Progress, 33(2), 490–499. doi:
Hunter et al.
and efficiency. Advances in Biochemical En- 10.1002/btpr.2418.
22 of 28
Current Protocols in Protein Science
Goodwin, E. C., & Rottman, F. M. (1992). The 3 - ment. Biotechnology Progress, 26(5), 1400–
flanking sequence of the bovine growth hormone 1410. doi: 10.1002/btpr.436.
gene contains novel elements required for effi- Imai-Nishiya, H., Mori, K., Inoue, M., Wak-
cient and accurate polyadenylation. Journal of itani, M., Iida, S., Shitara, K., & Satoh,
Biological Chemistry, 267(23), 16330–16334. M. (2007). Double knockdown of alpha1,6-
Graham, F. L., Smiley, J., Russell, W. C., & Nairn, fucosyltransferase (FUT8) and GDP-mannose
R. (1977). Characteristics of a human cell line 4,6-dehydratase (GMD) in antibody-producing
transformed by DNA from human adenovirus cells: A new strategy for generating fully
type 5. Journal of General Virology, 36(1), 59– non-fucosylated therapeutic antibodies with en-
74. doi: 10.1099/0022-1317-36-1-59. hanced ADCC. BMC Biotechnol, 7, 84. doi:
Graham, F. L., & van der Eb, A. J. (1973). A new 10.1186/1472-6750-7-84.
technique for the assay of infectivity of human Irfan, K., Rees, B., Barrett, T., Markice-
adenovirus 5 DNA. Virology, 52(2), 456–467. vic, B., & Segarra, C. (2015). Character-
doi: 10.1016/0042-6822(73)90341-3. ization and performance of the AllegroTM
Gustafsson, C., Minshull, J., Govindarajan, S., STR 1000 single-use stirred tank bioreactor
Ness, J., Villalobos, A., & Welch, M. (2012). system. BMC Proceedings, 9(9), P60. doi:
Engineering genes for predictable protein ex- 10.1186/1753-6561-9-s9-p60.
pression. Protein Expression and Purification, Jager, V., Bussow, K., Wagner, A., Weber, S.,
83(1), 37–46. doi: 10.1016/j.pep.2012.02.013. Hust, M., Frenzel, A., & Schirrmann, T. (2013).
Gutierrez-Granados, S., Cervera, L., Kamen, A. A., High level transient production of recombi-
& Godia, F. (2018). Advancements in mam- nant antibodies and antibody fusion proteins in
malian cell transient gene expression (TGE) HEK293 cells. BMC Biotechnology, 13, 52. doi:
technology for accelerated production of biolog- 10.1186/1472-6750-13-52.
ics. Critical Reviews in Biotechnology, (38)1– Janakiraman, V., Kwiatkowski, C., Kshirsagar, R.,
23. doi: 10.1080/07388551.2017.1419459. Ryll, T., & Huang, Y. M. (2015). Application
Hacker, D. L., Kiseljak, D., Rajendra, Y., Thurn- of high-throughput mini-bioreactor system for
heer, S., Baldi, L., & Wurm, F. M. (2013). systematic scale-down modeling, process char-
Polyethyleneimine-based transient gene expres- acterization, and control strategy development.
sion processes for suspension-adapted HEK- Biotechnology Progress, 31(6), 1623–1632. doi:
293E and CHO-DG44 cells. Protein Expres- 10.1002/btpr.2162.
sion and Purification, 92(1), 67–76. doi: Jayapal, K. P., Wlaschin, K. F., Hu, W. S., & Yap,
10.1016/j.pep.2013.09.001. M. G. S. (2007). Recombinant protein therapeu-
Halff, E. F., Versteeg, M., Brondijk, T. H., & tics from CHO cells - 20 years and counting.
Huizinga, E. G. (2014). When less becomes Chemical Engineering Progress, 103, 40–47.
more: Optimization of protein expression in Jinek, M., East, A., Cheng, A., Lin, S., Ma, E., &
HEK293-EBNA1 cells using plasmid titra- Doudna, J. (2013). RNA-programmed genome
tion - a case study for NLRs. Protein Ex- editing in human cells. Elife, 2, e00471 doi:
pression and Purification, 99, 27–34. doi: 10.7554/eLife.00471.
10.1016/j.pep.2014.03.010. Johnson, S., Oliver, C., Prince, G. A., Hemming,
Hasegawa, H., Woods, C. E., Kinderman, F., He, V. G., Pfarr, D. S., Wang, S. C., . . . Young, J.
F., & Lim, A. C. (2014). Russell body phe- F. (1997). Development of a humanized mono-
notype is preferentially induced by IgG mAb clonal antibody (MEDI-493) with potent in vitro
clones with high intrinsic condensation propen- and in vivo activity against respiratory syncytial
sity: Relations between the biosynthetic events virus. Journal of Infectious Diseases, 176(5),
in the ER and solution behaviors in vitro. MAbs, 1215–1224. doi: 10.1086/514115.
6(6), 1518–1532. doi: 10.4161/mabs.36242. Jones, D., Kroos, N., Anema, R., van Mont-
Hopkins, R. F., Wall, V. E., & Esposito, D. fort, B., Vooys, A., van der Kraats, S., . . .
(2012). Optimizing transient recombinant pro- Bout, A. (2003). High-level expression of re-
tein expression in mammalian cells. Meth- combinant IgG in the human cell line per.c6.
ods in Molecular Biology, 801, 251–268. doi: Biotechnology Progress, 19(1), 163–168. doi:
10.1007/978-1-61779-352-3_16. 10.1021/bp025574h.
Howorth, P. J. (1975). The physiological assess- Jordan, M., Kohne, C., & Wurm, F. M. (1998).
ment of acid-base balance. British Journal of Calcium-phosphate mediated DNA transfer into
Diseases of the Chest, 69(2), 75–102. doi: HEK-293 cells in suspension: Control of physic-
10.1016/0007-0971(75)90061-3. ochemical parameters allows transfection in
Hu, X., Xiao, C., Huang, Z., Guo, Z., Zhang, stirred media. Transfection and protein ex-
Z., & Li, Z. (2000). Pilot production of pression in mammalian cells. Cytotechnology,
u-PA with porous microcarrier cell cul- 26(1), 39–47. doi: 10.1023/A:10079173181
ture. Cytotechnology, 33(1-3), 13–19. doi: 81.
10.1023/A:1008127310890. Kao, F. T., & Puck, T. T. (1968). Genetics of so-
Huang, Y. M., Hu, W., Rustandi, E., Chang, K., matic mammalian cells, VII. Induction and iso-
Yusuf-Makagiansar, H., & Ryll, T. (2010). Max- lation of nutritional mutants in Chinese hamster
imizing productivity of CHO cell-based fed- cells. Proceedings of the National Academy of
batch culture using chemically defined media Sciences of the United States of America, 60(4),
1275–1281. doi: 10.1073/pnas.60.4.1275. Hunter et al.
conditions and typical manufacturing equip-
23 of 28
Current Protocols in Protein Science
Kaufman, R. J. (2000). Overview of vector de- Trends Biotechnology, 24(3), 137–142. doi:
sign for mammalian gene expression. Molec- 10.1016/j.tibtech.2006.01.007.
ular Biotechnology, 16(2), 151–160. doi: Kyriakopoulos, S., & Kontoravdi, C. (2014). A
10.1385/MB:16:2:151. framework for the systematic design of fed-
Kaufman, R. J., & Sharp, P. A. (1982). Am- batch strategies in mammalian cell culture.
plification and expression of sequences co- Biotechnology and Bioengineering, 111(12),
transfected with a modular dihydrofolate re- 2466–2476. doi: 10.1002/bit.25319.
ductase complementary dna gene. Journal Lacy-Hulbert, A., Thomas, R., Li, X. P., Lilley, C.
of Molecular Biology, 159(4), 601–621. doi: E., Coffin, R. S., & Roes, J. (2001). Interruption
10.1016/0022-2836(82)90103-6. of coding sequences by heterologous introns can
Kawabe, Y., Inao, T., Komatsu, S., Huang, G., enhance the functional expression of recombi-
Ito, A., Omasa, T., & Kamihira, M. (2017). nant genes. Gene Therapy, 8(8), 649–653. doi:
Improved recombinant antibody production by 10.1038/sj.gt.3301440.
CHO cells using a production enhancer DNA Lai, T., Yang, Y., & Ng, S. K. (2013). Ad-
element with repeated transgene integration at vances in Mammalian cell line development
a predetermined chromosomal site. Journal of technologies for recombinant protein produc-
Bioscience and Bioengineering, 123(3), 390– tion. Pharmaceuticals (Basel), 6(5), 579–603.
397. doi: 10.1016/j.jbiosc.2016.10.011. doi: 10.3390/ph6050579.
Keen, M. J., & Rapson, N. T. (1995). De- Lanoix, J., & Acheson, N. H. (1988). A rabbit beta-
velopment of a serum-free culture medium globin polyadenylation signal directs efficient
for the large scale production of recombi- termination of transcription of polyomavirus
nant protein from a Chinese hamster ovary DNA. EMBO Journal, 7(8), 2515–2522.
cell line. Cytotechnology, 17(3), 153–163. doi:
10.1007/BF00749653. Lanza, A. M., Kim, D. S., & Alper, H. S. (2013).
Evaluating the influence of selection markers on
Kim, T. K., & Eberwine, J. H. (2010). Mammalian obtaining selected pools and stable cell lines in
cell transfection: The present and the future. An- human cells. Biotechnology Journal, 8(7), 811–
alytical and Bioanalytical Chemistry, 397(8), 821.doi: 10.1002/biot.201200364.
3173–3178. doi: 10.1007/s00216-010-3821-6.
Lavner, Y., & Kotlar, D. (2005). Codon bias as a
Klein, R., Ruttkowski, B., Knapp, E., Salmons, factor in regulating expression via translation
B., Gunzburg, W. H., & Hohenadl, C. (2006). rate in the human genome. Gene, 345(1), 127–
WPRE-mediated enhancement of gene expres- 138. doi: 10.1016/j.gene.2004.11.035.
sion is promoter and cell line specific. Gene,
372, 153–161. doi: 10.1016/j.gene.2005.12.018. Le Fourn, V., Girod, P. A., Buceta, M., Regamey,
A., & Mermod, N. (2014). CHO cell en-
Knox, R., Nettleship, J. E., Chang, V. T., Hui, Z. gineering to prevent polypeptide aggrega-
K., Santos, A. M., Rahman, N., . . . Davis, tion and improve therapeutic protein secre-
S. J. (2013). A streamlined implementation of tion. Metabolic Engineering, 21, 91–102. doi:
the glutamine synthetase-based protein expres- 10.1016/j.ymben.2012.12.003.
sion system. BMC Biotechnology, 13, 74. doi:
10.1186/1472-6750-13-74. Lebkowski, J. S., Clancy, S., & Calos, M. P. (1985).
Simian virus 40 replication in adenovirus-
Kober, L., Zehe, C., & Bode, J. (2013). Opti- transformed human cells antagonizes gene ex-
mized signal peptides for the development of pression. Nature, 317(6033), 169–171. doi:
high expressing CHO cell lines. Biotechnology 10.1038/317169a0.
and Bioengineering, 110(4), 1164–1173. doi:
10.1002/bit.24776. Lee, J. S., Grav, L. M., Lewis, N. E., &
Faustrup Kildegaard, H. (2015). CRISPR/Cas9-
Koller, M. R., Hanania, E. G., Stevens, J., Eis- mediated genome engineering of CHO cell
feld, T. M., Sasaki, G. C., Fieck, A., & factories: Application and perspectives.
Palsson, B. O. (2004). High-throughput laser- Biotechnology Journal, 10(7), 979–994. doi:
mediated in situ cell purification with high pu- 10.1002/biot.201500082.
rity and yield. Cytometry A, 61(2), 153–161. doi:
10.1002/cyto.a.20079. Lee, J. S., Kallehauge, T. B., Pedersen, L.
E., & Kildegaard, H. F. (2015). Site-
Konno, Y., Kobayashi, Y., Takahashi, K., Taka- specific integration in CHO cells mediated by
hashi, E., Sakae, S., Wakitani, M., . . . Hosoi, S. CRISPR/Cas9 and homology-directed DNA re-
(2012). Fucose content of monoclonal antibod- pair pathway. Science Reports, 5, 8572. doi:
ies can be controlled by culture medium osmo- 10.1038/srep08572.
lality for high antibody-dependent cellular cyto-
toxicity. Cytotechnology, 64(3), 249–265. doi: Legmann, R., Schreyer, H. B., Combs, R. G., Mc-
10.1007/s10616-011-9377-2. Cormick, E. L., Russo, A. P., & Rodgers, S.
T. (2009). A predictive high-throughput scale-
Kumar, M., Keller, B., Makalou, N., & Sut- down model of monoclonal antibody production
ton, R. E. (2001). Systematic determination of in CHO cells. Biotechnology and Bioengineer-
the packaging limit of lentiviral vectors. Hu- ing, 104(6), 1107–1120. doi: 10.1002/bit.22474.
man Gene Therapy, 12(15), 1893–1905. doi:
10.1089/104303401753153947. Li, J., Menzel, C., Meier, D., Zhang, C., Dubel, S.,
& Jostock, T. (2007). A comparative study of
Kwaks, T. H., & Otte, A. P. (2006). Employ- different vector designs for the mammalian ex-
ing epigenetics to augment the expression pression of recombinant IgG antibodies. Journal
Hunter et al.
of therapeutic proteins in mammalian cells.
24 of 28
Current Protocols in Protein Science
of Immunological Methods, 318(1-2), 113–124. Matasci, M., Baldi, L., Hacker, D. L., & Wurm, F.
doi: 10.1016/j.jim.2006.10.010. M. (2011). The PiggyBac transposon enhances
Lin, N., Mascarenhas, J., Sealover, N. R., George, the frequency of CHO stable cell line gener-
H. J., Brooks, J., Kayser, K. J., . . . Archer- ation and yields recombinant lines with supe-
Hartmann, S. (2015). Chinese hamster ovary rior productivity and stability. Biotechnology
(CHO) host cell engineering to increase sia- and Bioengineering, 108(9), 2141–2150. doi:
lylation of recombinant therapeutic proteins 10.1002/bit.23167.
by modulating sialyltransferase expression. Mates, L., Chuah, M. K., Belay, E., Jerchow, B.,
Biotechnology Progress, 31(2), 334–346. doi: Manoj, N., Acosta-Sanchez, A., . . . Izsvak,
10.1002/btpr.2038. Z. (2009). Molecular evolution of a novel hy-
Liu, B., Spearman, M., Doering, J., Lattova, E., Per- peractive sleeping beauty transposase enables
reault, H., & Butler, M. (2014). The availability robust stable gene transfer in vertebrates. Na-
of glucose to CHO cells affects the intracellu- ture Genetics, 41(6), 753–761. doi: 10.1038/ng.
lar lipid-linked oligosaccharide distribution, site 343.
occupancy and the N-glycosylation profile of McCoy, R. E., Costa, N. A., & Morris, A. E. (2015).
a monoclonal antibody. Journal of Biotechnol- Factors that determine stability of highly con-
ogy, 170, 17–27. doi: 10.1016/j.jbiotec.2013.11. centrated chemically defined production media.
007. Biotechnology Progress, 31(2), 493–502. doi:
Liu, C., Dalby, B., Chen, W., Kilzer, J. M., & 10.1002/btpr.2047.
Chiou, H. C. (2008). Transient transfection fac- McCutchan, J. H., & Pagano, J. S. (1968). En-
tors for high-level recombinant protein produc- chancement of the infectivity of simian virus 40
tion in suspension cultured mammalian cells. deoxyribonucleic acid with diethylaminoethyl-
Molecular Biotechnology, 39(2), 141–153. doi: dextran. Journal of the National Cancer Insti-
10.1007/s12033-008-9051-x. tute, 41(2), 351–357.
Longo, P. A., Kavran, J. M., Kim, M. S., & Leahy, Melcher, R., Grosch, H. W., & Hasilik, A. (2002).
D. J. (2013). Generating mammalian stable Plasmid vectors with a 5 -hybrid intron facili-
cell lines by electroporation. Methods Enzymo- tate high-level glycoprotein expression in CHO-
logy, 529, 209–226. doi: 10.1016/B978-0-12-4 cells. Biochim Biophys Acta, 1575(1-3), 49–53.
18687-3.00017-3. doi: 10.1016/S0167-4781(02)00242-7.
Magistrelli, G., Malinge, P., Lissilaa, R., Fagete, S., Meuwly, F., Ruffieux, P. A., Kadouri, A.,
Guilhot, F., Moine, V., . . . Fischer, N. (2010). & von Stockar, U. (2007). Packed-bed
Rapid, simple and high yield production of bioreactors for mammalian cell culture:
recombinant proteins in mammalian cells us- Bioprocess and biomedical applications.
ing a versatile episomal system. Protein Ex- Biotechnology Advances, 25(1), 45–56. doi:
pression and Purification, 72(2), 209–216. doi: 10.1016/j.biotechadv.2006.08.004.
10.1016/j.pep.2010.04.007. Mimura, Y., Lund, J., Church, S., Dong, S., Li,
Mali, P., Yang, L., Esvelt, K. M., Aach, J., Guell, M., J., Goodall, M., & Jefferis, R. (2001). Bu-
DiCarlo, J. E., . . . Church, G. M. (2013). RNA- tyrate increases production of human chimeric
guided human genome engineering via Cas9. IgG in CHO-K1 cells whilst maintaining func-
Science, 339(6121), 823–826. doi: 10.1126/sci- tion and glycoform profile. Journal of Im-
ence.1232033. munological Methods, 247(1-2), 205–216. doi:
Mandenius, C. F., & Brundin, A. (2008). Biopro- 10.1016/S0022-1759(00)00308-2.
cess optimization using design-of-experiments Misaghi, S., Shaw, D., Louie, S., Nava, A., Sim-
methodology. Biotechnology Progress, 24(6), mons, L., Snedecor, B., . . . Cupp, J. E. (2016).
1191–1203. doi: 10.1002/btpr.67. Slashing the timelines: Opting to generate high-
Mangalampalli, V. R., Wycuff, D., Chen, M., titer clonal lines faster via viability-based sin-
Berlinger, D., Scheideman, E. H., Menon, gle cell sorting. Biotechnology Progress, 32(1),
A., . . . Schwartz, R. M. (2015). CHO-DHFR 198–207. doi: 10.1002/btpr.2204.
cell line development platform: Application Morgan, J. F., Morton, H. J., & Parker, R. C.
of Clonepix and Automated Mini Bioreactor (1950). Nutrition of animal cells in tissue cul-
(AMBR) technologies to meet accelerated time- ture; initial studies on a synthetic medium.
lines. BMC Proceedings, 9(Suppl 9), P52. doi: Proceedings of the Society for Experimen-
10.1186/1753-6561-9-S9-P52. tal Biology and Medicine, 73(1), 1–8. doi:
Mariati, Ho, S. C. L., Yap, M. G. S., & Yang, Y. 10.3181/00379727-73-17557.
(2010). Evaluating post-transcriptional regula- Mosser, M., Chevalot, I., Olmos, E., Blan-
tory elements for enhancing transient gene ex- chard, F., Kapel, R., Oriol, E., . . . Marc,
pression levels in CHO K1 and HEK293 cells. A. (2013). Combination of yeast hydrolysates
Protein Expression and Purification, 69(1), 9– to improve CHO cell growth and IgG pro-
15. doi: 10.1016/j.pep.2009.08.010. duction. Cytotechnology, 65(4), 629–641. doi:
Matasci, M., Bachmann, V., Baldi, L., Hacker, 10.1007/s10616-012-9519-1.
D. L., De Jesus, M., & Wurm, F. M. Muller, N., Girard, P., Hacker, D. L., Jordan, M.,
(2011). Rapid recombinant protein produc- & Wurm, F. M. (2005). Orbital shaker technol-
tion from pools of transposon-generated CHO ogy for the cultivation of mammalian cells in
cells. BMC Proceedings, 5(Suppl 8), P34. doi: suspension. Biotechnology and Bioengineering,
10.1186/1753-6561-5-S8-P34. 89(4), 400–406. doi: 10.1002/bit.20358. Hunter et al.

25 of 28
Current Protocols in Protein Science
Neville, J. J., Orlando, J., Mann, K., McCloskey, cells. Journal of Biotechnology, 153(1-2), 22–
B., & Antoniou, M. N. (2017). Ubiquitous 26. doi: 10.1016/j.jbiotec.2011.03.001.
Chromatin-opening Elements (UCOEs): Appli- Rajendra, Y., Kiseljak, D., Manoli, S., Baldi,
cations in biomanufacturing and gene therapy. L., Hacker, D. L., & Wurm, F. M. (2012).
Biotechnology Advances, 35(5), 557–564. doi: Role of non-specific DNA in reducing coding
10.1016/j.biotechadv.2017.05.004. DNA requirement for transient gene expression
Oliva-Trastoy, M., Defais, M., & Larminat, F. with CHO and HEK-293E cells. Biotechnology
(2005). Resistance to the antibiotic Zeocin by and Bioengineering, 109(9), 2271–2278. doi:
stable expression of the Sh ble gene does not 10.1002/bit.24494.
fully suppress Zeocin-induced DNA cleavage in Reusch, D., & Tejada, M. L. (2015). Fc glycans
human cells. Mutagenesis, 20(2), 111–114. doi: of therapeutic antibodies as critical quality at-
10.1093/mutage/gei016. tributes. Glycobiology, 25(12), 1325–1334. doi:
Park, S., & Stephanopoulos, G. (1993). Packed bed 10.1093/glycob/cwv065.
bioreactor with porous ceramic beads for animal Rio, D. C., Clark, S. G., & Tjian, R. (1985). A mam-
cell culture. Biotechnology and Bioengineering, malian host-vector system that regulates expres-
41(1), 25–34. doi: 10.1002/bit.260410105. sion and amplification of transfected genes by
Pazos, P., Boveri, M., Gennari, A., Casado, J., Fer- temperature induction. Science, 227(4682), 23–
nandez, F., & Prieto, P. (2004). Culturing cells 28. doi: 10.1126/science.2981116.
without serum: Lessons learnt using molecules Rocha-Pizana, M. D., Ascencio-Favela, G., Soto-
of plant origin. ALTEX, 21(2), 67–72. Garcia, B. M., Martinez-Fierro, M. L., & Al-
Peakman, T. C., Worden, J., Harris, R. H., Cooper, varez, M. M. (2017). Evaluation of changes in
H., Tite, J., Page, M. J., . . . Brett, S. (1994). promoters, use of UCOES and chain order to
Comparison of expression of a humanized improve the antibody production in CHO cells.
monoclonal antibody in mouse NSO myeloma Protein Expression and Purification, 132, 108–
cells and Chinese hamster ovary cells. Hum 115. doi: 10.1016/j.pep.2017.01.014.
Antibodies Hybridomas, 5(1–2), 65–74. doi: Rothem, L., Berman, B., Stark, M., Jansen,
10.3233/HAB-1994-51-209. G., & Assaraf, Y. G. (2005). The reduced
Plotkin, J. B., & Kudla, G. (2011). Synonymous folate carrier gene is a novel selectable
but not the same: The causes and consequences marker for recombinant protein overexpression.
of codon bias. Nature Reviews Genetics, 12(1), Molecular Pharmacology, 68(3), 616–624. doi:
32–42. doi: 10.1038/nrg2899. 10.1124/mol.105.013540.
Pollock, J., Ho, S. V., & Farid, S. S. (2013). Saunders, F., Sweeney, B., Antoniou, M. N.,
Fed-batch and perfusion culture processes: Stephens, P., & Cain, K. (2015). Chromatin
Economic, environmental, and operational function modifying elements in an industrial
feasibility under uncertainty. Biotechnology antibody production platform–comparison of
and Bioengineering, 110(1), 206–219. doi: UCOE, MAR, STAR and cHS4 elements.
10.1002/bit.24608. PLoS One, 10(4), e0120096. doi: 10.1371/
Preuss, A. K., Connor, J. A., & Vogel, H. (2000). journal.pone.0120096.
Transient transfection induces different intracel- Schek, N., Cooke, C., & Alwine, J. C. (1992).
lular calcium signaling in CHO K1 versus HEK Definition of the upstream efficiency element
293 cells. Cytotechnology, 33(1–3), 139–145. of the simian virus 40 late polyadenylation
doi: 10.1023/A:1008150402616. signal by using in vitro analyses. Molecular
Prevato, M., Ferlenghi, I., Bonci, A., Uematsu, Y., and Cellular Biology, 12(12), 5386–5393. doi:
Anselmi, G., Giusti, F., . . . Cozzi, R. (2015). 10.1128/MCB.12.12.5386.
Expression and characterization of recombinant, Sealover, N. R., Davis, A. M., Brooks, J. K.,
tetrameric and enzymatically active influenza George, H. J., Kayser, K. J., & Lin, N.
neuraminidase for the setup of an enzyme-linked (2013). Engineering Chinese hamster ovary
lectin-based assay. PLoS One, 10(8), e0135474. (CHO) cells for producing recombinant pro-
doi: 10.1371/journal.pone.0135474. teins with simple glycoforms by zinc-finger
Qian, Y., Khattak, S. F., Xing, Z., He, A., Kayne, nuclease (ZFN)-mediated gene knockout of
P. S., Qian, N. X., . . . Li, Z. J. (2011). Cell mannosyl (alpha-1,3-)-glycoprotein beta-1,2-N-
culture and gene transcription effects of cop- acetylglucosaminyltransferase (Mgat1). Jour-
per sulfate on Chinese hamster ovary cells. nal of Biotechnology, 167(1), 24–32. doi:
Biotechnology Progress, 27(4), 1190–1194. doi: 10.1016/j.jbiotec.2013.06.006.
10.1002/btpr.630. Sharp, P. M., & Li, W. H. (1987). The codon Adapta-
Rajendra, Y., Balasubramanian, S., Kiseljak, D., tion Index–a measure of directional synonymous
Baldi, L., Wurm, F. M., & Hacker, D. L. codon usage bias, and its potential applications.
(2015). Enhanced plasmid DNA utilization Nucleic Acids Research, 15(3), 1281–1295. doi:
in transiently transfected CHO-DG44 cells in 10.1093/nar/15.3.1281.
the presence of polar solvents. Biotechnology Shigekawa, K., & Dower, W. J. (1988). Electropo-
Progress, 31(6), 1571–1578. doi: 10.1002/btpr. ration of eukaryotes and prokaryotes: A general
2152. approach to the introduction of macromolecules
Rajendra, Y., Kiseljak, D., Baldi, L., Hacker, D. L., into cells. Biotechniques, 6(8), 742–751.
& Wurm, F. M. (2011). A simple high-yielding Shinkawa, T., Nakamura, K., Yamane, N.,
Hunter et al.
process for transient gene expression in CHO Shoji-Hosaka, E., Kanda, Y., Sakurada,
26 of 28
Current Protocols in Protein Science
M., . . . Shitara, K. (2003). The ab- Treschow, A., Unger, C., Aints, A., Felldin, U.,
sence of fucose but not the presence of Aschan, J., & Dilber, M. S. (2007). OuaSe-
galactose or bisecting N-acetylglucosamine lect, a novel ouabain-resistant human marker
of human IgG1 complex-type oligosac- gene that allows efficient cell selection within
charides shows the critical role of en- 48 h. Gene Therapy, 14(22), 1564–1572. doi:
hancing antibody-dependent cellular cyto- 10.1038/sj.gt.3303015.
toxicity. Journal of Biological Chemistry, Umana, P., Jean-Mairet, J., Moudry, R., Amstutz,
278(5), 3466–3473. doi: 10.1074/jbc.M210665 H., & Bailey, J. E. (1999). Engineered glyco-
200. forms of an antineuroblastoma IgG1 with op-
Singh, V. (1999). Disposable bioreactor for timized antibody-dependent cellular cytotoxic
cell culture using wave-induced agitation. activity. Nature Biotechnology, 17(2), 176–180.
Cytotechnology, 30(1–3), 149–158. doi: doi: 10.1038/6179.
10.1023/A:1008025016272. Vaheri, A., & Pagano, J. S. (1965). Infec-
Smelko, J. P., Wiltberger, K. R., Hickman, E. F., tious poliovirus RNA: A sensitive method
Morris, B. J., Blackburn, T. J., & Ryll, T. (2011). of assay. Virology, 27(3), 434–436. doi:
Performance of high intensity fed-batch mam- 10.1016/0042-6822(65)90126-1.
malian cell cultures in disposable bioreactor Villalobos, A., Ness, J. E., Gustafsson, C., Minshull,
systems. Biotechnology Progress, 27(5), 1358– J., & Govindarajan, S. (2006). Gene Designer: A
1364. doi: 10.1002/btpr.634. synthetic biology tool for constructing artificial
St Amand, M. M., Radhakrishnan, D., Robinson, A. DNA segments. BMC Bioinformatics, 7, 285.
S., & Ogunnaike, B. A. (2014). Identification of doi: 10.1186/1471-2105-7-285.
manipulated variables for a glycosylation con- Vishwanathan, N., Le, H., Jacob, N. M., Tsao, Y.
trol strategy. Biotechnology and Bioengineering, S., Ng, S. W., Loo, B., . . . Hu, W. S. (2014).
111(10), 1957–1970. doi: 10.1002/bit.25251. Transcriptome dynamics of transgene amplifica-
Stacey, G., & Davis, J. (2007). Medicines from ani- tion in Chinese hamster ovary cells. Biotechnol-
mal cell culture. Chichester; Hoboken, N.J.: Wi- ogy and Bioengineering, 111(3), 518–528. doi:
ley. 10.1002/bit.25117.
Stanley, P. (1983). Lectin-resistant CHO cells: von Horsten, H. H., Ogorek, C., Blanchard,
Selection of new mutant phenotypes. So- V., Demmler, C., Giese, C., Winkler, K.,
matic Cell Genetics, 9(5), 593–608. doi: . . . Sandig, V. (2010). Production of non-
10.1007/BF01574260. fucosylated antibodies by co-expression of het-
Stern, B., L. C., O., Trobe, C., Ravneberg, H., & erologous GDP-6-deoxy-D-lyxo-4-hexulose re-
Pryme, I. F. (2007). Improving mammalian cell ductase. Glycobiology, 20(12), 1607–1618. doi:
factories: The selection of signal peptide has a 10.1093/glycob/cwq109.
major impact on recombinant protein synthesis Wang, G., Zhang, W., Jacklin, C., Freedman, D.,
and secretion in mammalian cells. Trends in Cell Eppstein, L., & Kadouri, A. (1992). Modified
& Molecular Biology, 2, 1–7. CelliGen-packed bed bioreactors for hybridoma
Stettler, M., Zhang, X., Hacker, D. L., De Jesus, cell cultures. Cytotechnology, 9(1-3), 41–49.
M., & Wurm, F. M. (2007). Novel orbital shake doi: 10.1007/BF02521730.
bioreactors for transient production of CHO Welch, M., Villalobos, A., Gustafsson, C., & Min-
derived IgGs. Biotechnology Progress, 23(6), shull, J. (2009). You’re one in a googol: Op-
1340–1346. doi: 10.1021/bp070219i. timizing genes for protein expression. Jour-
Sun, X., Goh, P. E., Wong, K. T., Mori, T., nal of The Royal Society Interface, 6(Suppl
& Yap, M. G. (2006). Enhancement of tran- 4), S467–476. doi: 10.1098/rsif.2008.0520.
sient gene expression by fed-batch culture focus.
of HEK 293 EBNA1 cells in suspension. Wickens, M., Anderson, P., & Jackson, R. J.
Biotechnology Letters, 28(11), 843–848. doi: (1997). Life and death in the cytoplasm: Mes-
10.1007/s10529-006-9010-1. sages from the 3 end. Current Opinion in Ge-
Swiech, K., Picanco-Castro, V., & Covas, D. T. netics and Development, 7(2), 220–232. doi:
(2012). Human cells: New platform for recombi- 10.1016/S0959-437X(97)80132-3.
nant therapeutic protein production. Protein Ex- Wippermann, A., Rupp, O., Brinkrolf, K., Hof-
pression and Purification, 84(1), 147–153. doi: frogge, R., & Noll, T. (2015). The DNA methyla-
10.1016/j.pep.2012.04.023. tion landscape of Chinese hamster ovary (CHO)
Taki, S., Kamada, H., Inoue, M., Nagano, K., DP-12 cells. Journal of Biotechnology, 199, 38–
Mukai, Y., Higashisaka, K., . . . Tsunoda, S. 46. doi: 10.1016/j.jbiotec.2015.02.014.
(2015). A novel bispecific antibody against hu- Wurm, F., & Bernard, A. (1999). Large-scale tran-
man CD3 and ephrin receptor A10 for breast sient expression in mammalian cells for re-
cancer therapy. PLoS One, 10(12), e0144712. combinant protein production. Current Opin-
doi: 10.1371/journal.pone.0144712. ion in Biotechnology, 10(2), 156–159. doi:
Tchoudakova, A., Hensel, F., Murillo, A., Eng, 10.1016/S0958-1669(99)80027-5.
B., Foley, M., Smith, L., . . . Cacciuttolo, M. Wurm, F. M. (2004). Production of recombinant
(2009). High level expression of functional hu- protein therapeutics in cultivated mammalian
man IgMs in human PER.C6 cells. MAbs, 1(2), cells. Nature Biotechnology, 22(11), 1393–
163–171. doi: 10.4161/mabs.1.2.7945. 1398. doi: 10.1038/nbt1026.
Hunter et al.

27 of 28
Current Protocols in Protein Science
Xu, X., Nagarajan, H., Lewis, N. E., Pan, S., Yin, B., Gao, Y., Chung, C. Y., Yang, S., Blake,
Cai, Z., Liu, X., . . . Wang, J. (2011). The E., Stuczynski, M. C., . . . Betenbaugh, M.
genomic sequence of the Chinese hamster J. (2015). Glycoengineering of Chinese ham-
ovary (CHO)-K1 cell line. Nature Biotech- ster ovary cells for enhanced erythropoietin N-
nology, 29(8), 735–741. doi: 10.1038/nbt. glycan branching and sialylation. Biotechnology
1932. and Bioengineering, 112(11), 2343–2351. doi:
Yamane-Ohnuki, N., Kinoshita, S., Inoue-Urakubo, 10.1002/bit.25650.
M., Kusunoki, M., Iida, S., Nakano, R., . . . Zhou, H., Liu, Z. G., Sun, Z. W., Huang, Y.,
Satoh, M. (2004). Establishment of FUT8 & Yu, W. Y. (2010). Generation of sta-
knockout Chinese hamster ovary cells: An ideal ble cell lines by site-specific integration of
host cell line for producing completely defu- transgenes into engineered Chinese hamster
cosylated antibodies with enhanced antibody- ovary strains using an FLP-FRT system. Jour-
dependent cellular cytotoxicity. Biotechnol- nal of Biotechnology, 147(2), 122–129. doi:
ogy and Bioengineering, 87(5), 614–622. doi: 10.1016/j.jbiotec.2010.03.020.
10.1002/bit.20151. Zigler, J. S., Jr., Lepe-Zuniga, J. L., Vis-
Ye, J., Kober, V., Tellers, M., Naji, Z., Salmon, P., tica, B., & Gery, I. (1985). Analysis of the
& Markusen, J. F. (2009). High-level protein cytotoxic effects of light-exposed HEPES-
expression in scalable CHO transient transfec- containing culture medium. In Vitro Cellular
tion. Biotechnology and Bioengineering, 103(3), & Developmental Biology, 21(5), 282–287. doi:
542–551. doi: 10.1002/bit.22265. 10.1007/BF02620943.

Hunter et al.

28 of 28
Current Protocols in Protein Science

You might also like