Anda di halaman 1dari 22

Biotechnology Advances 29 (2011) 278299

Contents lists available at ScienceDirect

Biotechnology Advances
j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / b i o t e c h a d v

Research review paper

Towards high-yield production of pharmaceutical proteins with plant cell


suspension cultures
Jianfeng Xu a,b,, Xumeng Ge a, Maureen C. Dolan a
a
b

Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA


College of Agriculture and Technology, Arkansas State University, Jonesboro, AR 72401, USA

a r t i c l e

i n f o

Article history:
Received 25 July 2010
Received in revised form 24 December 2010
Accepted 2 January 2011
Available online 12 January 2011
Keywords:
Bioreactor
Bioprocess engineering
Plant cell culture
Protein production
Recombinant protein
Therapeutic protein

a b s t r a c t
Molecular farming in plants with signicant advantages in cost and safety is touted as a promising platform
for the production of complex pharmaceutical proteins. While whole-plant produced biopharmaceuticals
account for a signicant portion of the preclinical and clinical pipeline, plant cell suspension culture, which
integrates the merits of whole-plant systems with those of microbial fermentation, is emerging as a more
compliant alternative factory. However, low protein productivity remains a major obstacle that limits
extensive commercialization of plant cell bioproduction platform. This review highlights the advantages and
recent progress in plant cell culture technology and outlines viable strategies at both the biological and
process engineering levels for advancing the economic feasibility of plant cell-based protein production.
Approaches to overcome and solve the associated challenges of this culture system that include nonmammalian glycosylation and genetic instability will also be discussed.
2011 Elsevier Inc. All rights reserved.

Contents
1.
2.

3.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Plant cell cultures as alternative bioproduction platform for pharmaceutical
2.1.
Advantages over whole-plant system . . . . . . . . . . . . . . .
2.2.
Pharmaceutical proteins produced by plant cell cultures
. . . . .
2.2.1.
Antibodies . . . . . . . . . . . . . . . . . . . . . . .
2.2.2.
Vaccines . . . . . . . . . . . . . . . . . . . . . . . .
2.2.3.
Cytokines, growth factors and growth hormones . . . . .
2.2.4.
Therapeutic enzymes and others . . . . . . . . . . . . .
2.2.5.
Other pharmaceutical proteins . . . . . . . . . . . . .
Strategies for achieving high-yield production of pharmaceutical proteins .
3.1.
Molecular approaches for increased expression . . . . . . . . . .
3.1.1.
Enhancing gene transcription . . . . . . . . . . . . . .
3.1.2.
Improving translation efciency . . . . . . . . . . . . .
3.1.3.
Minimizing post-translational protein degradation . . . .
3.1.4.
Stabilizing protein fusions and Hyp-Glyco technology . . .
3.2.
Cell culture approaches . . . . . . . . . . . . . . . . . . . . .
3.2.1.
Higher order testing of culture medium . . . . . . . . .
3.2.2.
Cell immobilization . . . . . . . . . . . . . . . . . . .
3.2.3.
In situ protein removal . . . . . . . . . . . . . . . . .
3.3.
Bioreactor engineering approaches . . . . . . . . . . . . . . . .
3.3.1.
Optimized bioreactor design and operation . . . . . . .
3.3.2.
Advanced bioreactor culture strategies . . . . . . . . . .
3.3.3.
Adoption of disposable bioreactors . . . . . . . . . . .
3.4.
Downstream processing . . . . . . . . . . . . . . . . . . . . .

. . . .
proteins
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .
. . . .

. .
.
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .
. .

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

279
280
280
281
281
282
282
282
282
282
283
283
285
285
285
286
286
287
287
287
288
289
290
290

Corresponding author. Arkansas Biosciences Institute, Arkansas State University, P.O. Box 639, State University, AR 72467, USA. Tel.: + 1 870 680 4812; fax: + 1 870 972 2026.
E-mail address: jxu@astate.edu (J. Xu).
0734-9750/$ see front matter 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.biotechadv.2011.01.002

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

4.

Other challenges to overcome . . .


4.1.
Non-mammalian glycosylation
4.2.
Genetic instability . . . . . .
5.
Perspective and conclusion . . . . .
Acknowledgements . . . . . . . . . . .
References . . . . . . . . . . . . . . .

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

1. Introduction
Protein therapeutics have revolutionized modern medicine and
are currently in use for the treatment of many human diseases
including diabetes, anemia, hepatitis and cancer (Leader et al., 2008;
Woodnutt et al., 2008). To date, nearly 100 proteins with human
therapeutic applications have entered the market (Hacker et al., 2009)
and in excess of 370 are currently under development. Included
among this group of proteins are mainly vaccines, antibodies and
antibody derivatives in addition to several serum-derived proteins
including cytokines, growth hormones, interleukins, and interferon.
Worldwide protein drug sales currently estimated to be more than
$90 billion per year, are expected to reach $125 billion by the year
2015 (Decision Resources 2009 at http://www. decisionresources.
com/). Commercial production of such pharmaceutical proteins has
traditionally relied on bacterial fermentation or mammalian cellbased production, however, limitations including cost, scalability,
safety and protein authenticity with these expression systems have
prompted research into alternative platforms (Boehm, 2007; Streateld,
2007). Plant-based molecular farming has emerged as a promising
approach with signicant advantages in both cost and safety over
other eukaryotic expression systems (Basaran and Rodriguez-Cerezo,
2008; Doran, 2000; Fischer et al., 2004; Lau and Sun, 2009; Ma et al.,
2003; Sharma and Sharma, 2009; Streateld, 2007; Tremblay et al.,
2010). Fig. 1 compares several production characteristics of plant-based
production platforms with those of other systems when used for
pharmaceutical protein expression.
Molecular farming encompasses the use of either whole-plants or
in vitro cultured plant cell/tissues for the production of recombinant
proteins. While one of the seminal features of a whole-plant based

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

279

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

292
292
293
294
294
294

platform is providing a readily scalable system, this platform does


have several drawbacks including long development times for stable
transgenics, instability of product yield and quality, issues with
possible fertilizer contamination, impact of pests, diseases and
weather conditions on the production factory, and most importantly
challenges in applying good manufacturing practice (GMP) to the
whole-plant production pipeline (Hellwig et al., 2004; Karg and Kallio,
2009; Shih and Doran, 2009). Plant cell suspension culture integrates
many of the merits of the whole-plant system with those of microbial
and animal cell cultures (Hellwig et al., 2004), and holds tremendous
promise as a factory for valuable pharmaceutical proteins (Fig. 2).
Like the microbial cells, undifferentiated plant calli can be separated
and propagated under a controlled, liquid media environment to
generate stable cell suspension cultures. This culture technology
developed in the 1950s, has been primarily used for the production of
valuable secondary metabolites such as paclitaxel, shikonin, artemisinin, digoxin, ginsenosides and ajmalicine (Georgiev et al., 2009; Rao
and Ravishankar, 2002; Smetanska, 2008; Weathers et al., 2010; Xu et
al., 1998a). Commercial production of paclitaxel in Taxus cell
suspension cultures developed by Phyton Biotech (http://www.
phytonbiotech.com/) has provided Bristol-Myers Squibb with a
secure, sustainable and environmentally-friendly source of paclitaxel
for Taxol since 1995 (Roberts and Shuler, 1997). With considerable
advances in plant molecular biology over the subsequent 15 years, as
well as an ever increasing demand for protein therapeutics that meet
regulatory and safety standards, plant cell culture has emerged as a
viable alternative production platform for pharmaceutical proteins.
The benets of plant cell culture in achieving human-like protein
production have long been recognized. Like bacteria, plant cells have
relatively rapid doubling times and can be grown in simple synthetic

Fig. 1. Comparison of key production parameters of the different expression systems used for recombinant therapeutic proteins. Note: the purication cost of the plant cell culture
system will become low if recombinant proteins are secreted into medium.

280

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

Fig. 2. Schematic depicting the plant cell suspension culture process for producing recombinant proteins. Production plant cell lines can be established from either the transgenic
whole-plant (upper-pathway) or by direct transformation of cultured plant cells with the target genes (lower-pathway). Scale-up production and downstream purication
methodologies adapted from other commercialized cell-based bioproduction systems can be used in delivering plant-made pharmaceuticals and proteins to the marketplace.

media using conventional bioreactors. However like mammalian cells,


plants are eukaryotes and can perform nearly all post-translational
modications that occur in human cells. As such, plant cell culture is
considered comparable to conventional mammalian cell culture including
Chinese hamster ovary (CHO) cells, which are routinely used for the
production of complex protein therapeutics. However, the most salient
advantage the plant cell system has over its mammalian counterpart is
plant cells do not harbor any known human pathogens. This issue has
received signicant consideration and review after Genzyme experienced
interruption (June, 2009) in their CHO cell production of Cerezyme, the
commercial Gaucher disease therapeutic, due to infection of their
production cell line with a calicivirus (Rybicki, 2010). This serious safety
concern for human patients and an immediate need to ensure a
sustainable supply chain for this orphan drug highlights the importance
and urgency for continued efforts to improve the performance of plant cell
culture bioproduction system for therapeutic proteins in the future.
Of signicant relevance to this review, the world's largest drug
company, Pzer, recently acquired licensing rights from Protalix
Biotherapeutics (Israel) for their carrot cell-based production system
of glucocerebrosidase, an orphan drug for Gaucher disease (Aviezer
et al., 2009a, 2009b; Shaaltiel et al., 2007). This followed with Protalix
receiving a Phase III Fast Track Designation for this drug in August,
2009 from the U.S. Food and Drug Administration (FDA). The Pzer
licensing transaction, which exceeded $100 million, highlights the
commitment of big pharmaceutical companies and their condence in
the commercial viability of plant cell-based production for pharmaceutical proteins. This product was approved for use in the treatment
of Gaucher disease in France by the French regulatory authority in
July, 2010, thus becoming the rst plant-made human pharmaceutical
to enter the commercial sector. Approval by FDA for use of this drug in
the USA is expected to come in early 2011. Further illustrating the
sustainability of this production platform, Protalix announced in June,

2010 that a second protein candidate produced through plant cell culture
system, PRX-105 that is a human acetylcholinesterase for treatment of
nerve-gas and pesticide poisoning, has been completed Phase I clinical
trials, and another two plant cell-produced enzymes, PRX-12 (galactosidase enzyme replacement therapy for Fabry disease) and pr-antiTNF
(biosimilar version of Enbrel) are in its preclinical pipeline (http://www.
protalix.com). Protalix's landmark success in commercializing plant cellmade human proteins was in fact preceded by Dow AgroSciences who
received the world's rst regulatory approval by US Department of
Agriculture (USDA) for tobacco cell-based vaccine against Newcastle
disease virus in 2006. The commercialization success of plant cell
bioproduction by these two companies undeniably sets the stage for
entry of other plant-made pharmaceuticals into the marketplace.
The molecular farming industry is still in its early development
(Sharma and Sharma, 2009). While plant biotechnology continues to
drive increasing numbers of whole-plant produced biopharmaceuticals into the preclinical and clinical development pipeline (Karg and
Kallio, 2009), the plant cell culture system is clearly emerging as a
more immediate alternative plant-based factory for producing
complex pharmaceutical proteins (Hellwig et al., 2004; Shih and
Doran, 2009). This review highlights the recent progress of this
promising bioproduction platform for pharmaceutical proteins and
outlines a range of strategies taken or underway to increase the
protein yields from plant cell cultures.
2. Plant cell cultures as alternative bioproduction platform for
pharmaceutical proteins
2.1. Advantages over whole-plant system
In comparison to whole-plant cultivation, plant cell culture
platforms have several notable advantages including: 1) rapid growth

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

with cell doubling time as short as one day; 2) improved consistency


of protein product with the use of controlled bioreactors which are
less prone to biotic- and abiotic-induced variations that commonly
hamper eld/greenhouse grown plant-based protein production;
3) fewer regulatory and environmental compliance hurdles for ensuring product quality and safety and limited to no GMO (Genetically
Modied Organism) release issues and; 4) simple procedure for
secreted protein isolation/purication from inexpensive, well-dened
medium that lacks exogenous proteins which complicate the
purication process (Lee et al., 2000). Notably, plant suspension cell
cultures are composed of dedifferentiated cells that lack fully
functional plasmodesmata, and hence systemic post-transcriptional
gene silencing (PTGS) may be reduced since PTGS is generally
transmitted via plasmodesmata and the vascular system (Crawford
and Zambryski, 1999; Doran, 2000; Su and Lee, 2007). More detailed
discussions of the advantages of plant cell-based production platform
can be found in several recent reviews (Hellwig et al., 2004; Huang
and McDonald, 2009; Shih and Doran, 2009).
2.2. Pharmaceutical proteins produced by plant cell cultures
Following expression of the rst complex recombinant protein
(human serum albumin) in tobacco cell culture (Sijmons et al., 1990),
a diverse array of pharmaceutical proteins including antibodies,
vaccines, hormones, growth factors and cytokines have been
produced in plant cells with the majority having demonstrated
bioactivity (Kwon et al., 2003b; Lienard et al., 2007; Magnuson et al.,
1998; Matsumoto et al., 1993; Smith et al., 2002). Tobacco BY-2
(Bright yellow-2) and NT-1 (Nicotiana tabacum-1) cells are the most
frequently chosen host lines used for recombinant protein expression.
These closely related cell lines are fast growing (specic growth
rate up to 0.044 h 1), robust, and readily undergo Agrobacteriummediated transformation and cell cycle synchronization (Hellwig
et al., 2004; Ozawa and Takaiwa, 2010; Su and Lee, 2007; Tremblay
et al., 2010). Other plant hosts used for establishing cell cultures have
included rice (Huang et al., 2005; Torres et al., 1999), soybean (Smith
et al., 2002), alfalfa (Daniell and Edwards, 1995) and tomato (Kwon
et al., 2003a). Plant cell lines derived from common edible crop
species, may in fact be more favorable than tobacco cells in terms of
by-product levels and regulatory compliance (Hellwig et al., 2004). An

281

extensive list of recombinant pharmaceutical proteins expressed


using plant cell-based platforms has been recently reviewed by Huang
and McDonald (2009). Table 1 summarizes the pharmaceutical
proteins produced at high yields (N10 mg/L or N1% TSP) in plant cell
suspension cultures to date.
2.2.1. Antibodies
Antibodies have emerged as the dominant class of recombinant
proteins in the pharmaceutical industry (De Muynck et al., 2010).
Since expression of the rst full-size monoclonal antibody (mAb) in
tobacco over 20 years ago (Hiatt et al., 1989), plant-produced
antibodies (Plantibodies) have received considerable interest (Ko
et al., 2009; Russell and Fuller, 2000; Stoger et al., 2002a; Tavladoraki
et al., 1993). Assembly of a number of functional immunoglobulin G
(IgG, primarily IgG1) and immunoglobulin A (IgA) antibodies has
been demonstrated in Nicotiana, Arabidopsis and other plants (De
Jaeger et al., 2000; De Wilde et al., 1998; Lai et al., 2010; Larrick et al.,
2001; Ma et al., 2003; Ramirez et al., 2003; Tremblay et al., 2010). For
several of the complex molecular forms of immunoglobulins including
secretory IgA (sIgA) and IgG4, plants in fact offer the only
commercially viable system for large-scale production to date (De
Muynck et al., 2010; Ma et al., 2003; Shaaltiel et al., 2009; Stoger et al.,
2002a).
There is an increasing interest in plant cell-based production
platforms for functional mAbs due to the rising concern about
potential safety issues. Examples include the Guy's 13 IgG1 (Fischer
et al., 1999b; Sharp and Doran, 2001a, 2001b), a human mAb against
hepatitis B virus surface antigen (HBsAg) (Yano et al., 2004), a human
anti-rabies virus mAb (Girard et al., 2006), and most recently a human
anti-HIV mAb (Holland et al., 2010) all of which have been reported to
be expressed in tobacco cell suspension cultures. Challenges do
remain where completely assembled IgG antibodies while actively
secreted into the culture media and fully functional, were shown to
be susceptible to rapid proteolytic degradation (Sharp and Doran,
2001a). With the exception of full-size mAbs, plants or plant cells
have also been quite successful at expressing several antibody
derivatives of therapeutic value, including Fab fragments, singlechain Fvs (scFv), bispecic Fvs, diabodies, minibodies, and single
variable domains (Almquist et al., 2006; De Wilde et al., 1998; Ma
et al., 2003; Ramirez et al., 2002, 2000, 2001; Shaaltiel et al., 2009;

Table 1
High-yield expression of pharmaceutical proteins in plant cell suspension cultures.
Therapeutic proteins

Host cells

Protein yields

Promoter

Localization

Reference

Human interferon 2b (hIFN 2)


Human growth hormone (hGH)

N. tabacum cv BY-2
N. tabacum cv BY-2
O. sativa L. cv. Donjin
Glycine max cv Williams82 (Soybean)
N. tabacum cv BY-2
N. tabacum cv NT-1
O. sativa (rice)
O. sativa cv Taipei 309
O. sativa (rice)
O. sativa (rice)
O. sativa (rice)
O. sativa (rice)

28 mg/L
35 mg/L
57 mg/l
22 mg/L
15 mg/L
30 mg/L
200 mg/L
85 mg/L
247 mg/L
110 mg/L
50 mg/L
76.5 mg/L

CaMV35S
CaMV35S
RAmy3D
(ocs)3mas
CaMV35S
CaMV35S
RAmy3D
RAmy3D
RAmy3D
RAmy3D
RAmy3D
RAmy3D

Secreted
Secreted
Secreted
Intracellular
~ 50% secreted
Secreted
Secreted
Secreted
Secreted
Secreted
Secreted
Secreted

Xu et al. (2007)
Xu et al. (2010)
Kim et al. (2008a)
Smith et al. (2002)
Yano et al. (2004)
Francisco et al. (1997)
Huang et al. (2001)
Terashima et al. (1999)
McDonald et al. (2005)
Trexler et al. (2005)
Trexler et al. (2002)
Park et al. (2010)

O. sativa (rice)

129 mg/L

RAmy3D

Secreted

Shin et al. (2003)

O. sativa cv Taipei 309


O. sativa (rice)
O. sativa (rice)
N. tabacum cvNT-1
N. tabacum cvNT-1
N. tabacum cv. BY-2.
N. tabacum cv BY-2
Acanthopanax senticosus
N. tabacum cv BY-2

3%4% TSP
77 mg/L
31 mg/L
27 mg/L
10.8 mg/L
~ 10 mg/L
10 mg/L
3.6% TSP
4.3% TSP

RAmy3D
RAmy3D
RAmy3D
CaMV35S
CaMV35S
CaMV35S
CaMV35S
SWPA2
SWPA2

Intracellular
Secreted
Secreted
Secreted
Secreted
Secreted
Secreted
Intracellular
Intracellular

Huang et al. (2002b)


Huang et al. (2005)
Shin et al. (2010b)
Becerra-Arteaga et al. (2006)
Wongsamuth and Doran (1997)
Holland et al. (2010)
Fu et al. (2009)
Jo et al. (2006)
Choi et al. (2003)

Hepatitis B surface antigen (HBsAg)


mAb against HBsAg
Bryodin 1
Human 1-antitrypsin (rAAT)

Human cytotoxic T lymphocyte antigen


4-immunoglobulin (hCTLA4Ig)
Human granulocyte-macrophage colony
stimulating factor (hGM-CSF)
Human lysozyme
Human serum albumin (HSA)
Human interlukin-12 (IL-12)
Human alkaline phosphatase
Mouse IgG anti Streptococcus surface antigen
Human anti-HIV antibody 2G12
Human -iduronidase
Human lactoferrin
*TSP: total soluble proteins.

282

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

Tavladoraki et al., 1993). For example, an anti-phytochrome scFv


(Firek et al., 1994), a scFv against human spleen ferritin (Semenyuk
et al., 2003) and a mouse Fab against surface protein antigen of
Streptococcus mutants serotype c (Yano et al., 2006) have been
reported to express in tobacco cells with full function.
2.2.2. Vaccines
Plants have emerged as an attractive production system for
subunit vaccines which unlike traditional whole pathogen-based
vaccines, present only select immunogenic epitopes of the pathogen
for eliciting production of antibodies in the host organism (Chichester
et al., 2009; Daniell et al., 2009; Franconi et al., 2010; Rybicki, 2010;
Tiwari et al., 2009; Walter, 2008; Wu et al., 2009; Yu and Langridge,
2001). One of the unique features of plant-produced vaccines is
the concept that plants not only serve as the production system
but also as the delivery vehicle for oral vaccines (Tiwari et al., 2009;
Wang and Coppel, 2008). A complete list of vaccine antigens
expressed in plants can be seen in several recent reviews (Rybicki,
2010).
Plant cells offer most of the advantages of whole-plant system for
the expression of vaccines, along with controlled, sterile, reproducible
production that is aligned with GMP manufacturing requirements
(Tiwari et al., 2009). An example of a commercial plant cell platform is
the Concert Plant-Cell-Produced system developed by Dow AgroSciences for delivering vaccines to poultry and livestock (http://www.
dowagro.com/animalhealth/). Multiple antigens have been expressed
using this platform including heat-labile toxin from E. coli, hemagglutinin neuraminidase (HN), Newcastle disease virus (NDV), hemagglutinin (HA) from avian inuenza and VP2 from infectious bursal
disease (IBD). These plant-based vaccines have shown effective
immune responses in poultry and swine and provide a high-level
protection for animals challenged with the various disease pathogens
(Mihaliak and Webb, 2005). Plant-based platforms have garnered
particular interest for the production of the HBsAg antigen (Guan
et al., 2010). In addition to whole-plants, expression of the HBsAg has
been reported in both soybean and tobacco cell suspension cultures
(Ganapathi et al., 2007; Kumar et al., 2005, 2007; Sojikul et al., 2003).
Another example of a plant cell-produced vaccine targets the intimin
gene product of E. coli O157:H7 (Judge et al., 2004). Mice immunized
intraperitoneally with the recombinant intimin or fed with the
transgenic plant cells expressing this protein could generate an
intimin-specic mucosal immune response and exhibited a reduced
duration of E. coli O157:H7 fecal shedding following challenge (Judge
et al., 2004).
2.2.3. Cytokines, growth factors and growth hormones
Cytokines play a major role in orchestrating virtually every aspect
of the immune system, and thus have emerged as an interesting class
of promising immunotherapy agents (Cannon, 2000). As one of the
most important family of cytokines, bioactive interleukins of human
and animal origin have been produced in multiple plant species
including tobacco, tomato, rice and potato plant systems (GutierrezOrtega et al., 2004, 2005; Ma et al., 2005; Medrano et al., 2009, 2010;
Park and Cheong, 2002; Wang et al., 2008). In the context of plant cellbased expression, tobacco cells have been used to express bioactive
human interleukin-2 and -4 (hIL-2 and -4) (Magnuson et al., 1998),
human interleukin-18 (hIL-18) (Sharma et al., 2006), and human
interleukin-12 (hIL-12) (Kwon et al., 2003b). Of particular signicance to this review is the recent report of improved expression of
hIL-12 using a rice cell culture-based expression platform (Shin et al.,
2010b). Rice cells appear to exhibit lower associated proteolytic
activity on hIL-12 than all other plant-based platforms tested for
expression of this cytokine to date.
In addition to the interleukins, human granulocyte macrophage
colony stimulating factor (hGM-CSF) is another important cytokine
for which plants and plant cells have received much attention as a

production platform (James et al., 2000; Joo et al., 2006; Kwon et al.,
2003a, 2003c; Lee et al., 2004a; Shin et al., 2003). While very low
secreted protein yields of hGM-CSF (0.0070.07 mg/L) (Lee et al.,
2004b) were recovered when expressed in tobacco cells, high levels of
the protein (129 mg/L) were achieved using transgenic rice cell
expressed under an inducible rice amylase 3D (Ramy3D) promoter
(Shin et al., 2003). Conversely, functional human growth hormone
(hGH) has been expressed at high levels in both rice cells (57 mg/L)
(Kim et al., 2008a) and tobacco BY-2 cells (35 mg/L) (Xu et al., 2010).
Success of plant-based production may be protein specic however as
growth factors including human epidermal growth factor (hEGF)
(Parsons et al., 2010; Wirth et al., 2004) and insulin-like growth factor
(IGF-1) (Panahi et al., 2004, 2003) were produced at low protein
yields in plant cells.
2.2.4. Therapeutic enzymes and others
Several enzymes with therapeutic/diagnostic applications have
been produced in plant cell bioreactors. For example tobacco BY-2
cells have been shown to produce human tissue transglutaminase
(htTG), an enzyme used in the diagnosis of coeliac disease (Sorrentino
et al., 2005). Human lysozyme has been expressed in both rice cells
and rice grains for its potential utility as an antimicrobial food
supplement (Huang et al., 2002a, 2002b). In addition, several human
lysosomal enzymes have been produced in tobacco BY-2 cells
including human -iduronidase which has application as an enzyme
replacement therapy for treating mucopolysaccharidosis I (MPS I). Of
particular interest is the recombinant human glucocerebrosidase
enzyme (prGCD) that is being produced in carrot cells by Protalix
for commercial distribution in the treatment of Gaucher disease. The
carrot cell-produced prGCD naturally bears terminal mannose glycans
which eliminates the current requirement for post-production
chemical trimming of the sugars of the mammalian cell-expressed
recombinant GCD (sold under the trade name, Cerezyme). This new
prGCD is being developed for the human enzyme replacement
therapy market under the brand name of UPLYSO (Aviezer et al.,
2009a, 2009b; Shaaltiel et al., 2007). Preclinical and human clinical
trials of UPLYSO have shown the serum half-life of this plantexpressed GCD is signicantly longer than that of mammalian cellexpressed Cerezyme (http://www.protalix.com/). Other enzymes in
preclinical and clinical development using this plant cell platform
include -galactosidase for the treatment of Fabry disease; pr-antiTNF,
a biosimilar version of Enbrel and; acetylcholinesterase to treat nervegas and pesticide poisoning (Karg and Kallio, 2009).
2.2.5. Other pharmaceutical proteins
Other pharmaceutical proteins expressed in plant cell cultures
include dust mite allergens produced in tobacco BY-2 cell for use in
allergy diagnosis or immunotherapy (Lienard et al., 2007); a human
1-antitrypsin (AAT) produced in rice cell for treatment of hereditary
disorder in which a deciency of AAT leads to a chronic uninhibited
tissue breakdown (Terashima et al., 1999; Trexler et al., 2002, 2005); a
human lactoferrin produced in Siberian ginseng cell for use in advanced
nutritional products and in therapy for systemic infections; and a
thrombomodulin derivative produced in tobacco BY-2 cells for the
treatment of thrombotic disorders (Schinkel et al., 2005). As the
number of recombinant proteins shown to be successfully expressed
in plants increases, the production of pharmaceutical and diagnostic
proteins that leverage plant cell cultures as a commercially valid
platform is anticipated to gain broader acceptance and wider adoption.
3. Strategies for achieving high-yield production of
pharmaceutical proteins
While plant cell culture holds tremendous promise as an
alternative production platform for pharmaceutical proteins, several
bottlenecks limit the commercialization of this technology. Low

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

protein yields remain problematic; typically ranging from 0.01 to


200 mg/L (Huang and McDonald, 2009; James and Lee, 2001; Su and
Lee, 2007). Recent advances in plant molecular biology have greatly
improved protein yields well beyond the threshold of 10 mg/L that is
generally regarded as the requisite expression range for entry into
commercial process development (Hellwig et al., 2004). However
the need for additional research and development is necessary to
further boost the production levels in the order of 5- to 10-fold to
attain the desired prot margin for plant-based recombinant protein
expression. To reach this goal, a systematic strategy is required to
maximize the efciency of all stages of the production pipeline from
gene expression to cell culture, process development, and nally to
downstream protein purication (Ponstein et al., 1996).
In the past two decades, numerous approaches have been
exploited to enhance the production of heterologous protein in
plant cell cultures. Categorized based upon production stages, these
approaches can be classied into four groups that include molecular,
cell culture, process engineering (bioreactor culture) and downstream processing approaches (Fig. 3). It is necessary that these
approaches are properly integrated into the production pipeline to
achieve the desired commercial success of plant cell-based bioproduction of recombinant proteins. A similar approach has been
systematically adopted by the mammalian cell culture industry over
the past 20 years, leading to its high productivity today.

283

3.1. Molecular approaches for increased expression


The most critical strategies in enhancing the transgene expression
levels in plant cell culture systems incorporate molecular methodologies. These approaches target mainly the two genetic information
transfer processes dened by the central dogma: transcription and
translation (Desai et al., 2010; Sharma and Sharma, 2009; Streateld,
2007). Great strides have been made over the past two decades to
improve heterologous protein expression in plant cells through
enhanced gene transcription, proper mRNA processing, improved
translation efciency, and incorporation of novel protein fusion
technology (Desai et al., 2010; Ponstein et al., 1996; Streateld,
2007). In addition, improved post-translational protein stability can
play a pivotal rule in achieving high protein yields (Doran, 2006a).
3.1.1. Enhancing gene transcription
Increasing the transcription rate of stably transformed gene
sequences is the most direct and efcient approach for boosting
protein expression. This is best achieved by using a strong constitutive
or inducible promoter and understanding the promoter elements and
factors that can be used to modulate transgene transcription. In
general the goal is to generate target mRNA yields approaching 10
20% of the total mRNA (Hellwig et al., 2004). In the context of plant
cell culture, the characteristics of various promoters used for

Fig. 3. Systematic strategies to achieve high-yield protein production with plant cell suspension cultures.

284

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

expressing foreign genes in this system has been summarized by


Huang and McDonald, 2009. The major plant expression promoters
are also discussed extensively in recent reviews (Desai et al., 2010;
Sharma and Sharma, 2009), though the focus is placed on the wholeplant expression system.
3.1.1.1. Constitutive promoters. Constitutive promoters directly and
continually drive the expression of the transgene in all plant tissues
and are generally not dependent of developmental stage of the
production host. The best-known and most widely used constitutive
promoter in plant cell culture is derived from the Cauliower Mosaic
Virus (CaMV35S) and is theoretically capable of continuously driving
recombinant protein expression until the plant cells reach a
stationary phase in growth (Huang and McDonald, 2009). The
CaMV35S promoter sequence has been duplicated and/or modied
to further improve the gene transcription efciency up to 10-fold
(Fischer et al., 1999b; He et al., 2008; Kay et al., 1987). Alternative
constitutive promoters used in plant cell culture include the ubiquitin
promoter and the (ocs)3mas promoter (Hellwig et al., 2004). The
ubiquitin promoter, isolated from a variety of plants including maize,
Arabidopsis, potato, sunower, tobacco and rice (Sharma and Sharma,
2009), has been used to express scFv antibody and insulin-like
growth factor-1 (IGF-1) in rice cells (Panahi et al., 2004; Torres et al.,
1999). The hybrid (ocs)3mas promoter, constructed from octopine
synthase (ocs) and mannopine synthase (mas) promoter sequences,
was used for the expression of HBsAg in soybean cell culture, which
resulted in high levels of secreted protein on the order of 22 mg/L
(Smith et al., 2002). However constitutive expression is regarded as
a growth-associated process and thus may present signicant
metabolic burden on the plant cell during critical stages of cell
growth and thus a reduced specic growth rate. In addition, the
constant accumulation of the foreign product may create a toxic
environment that negatively impacts host cell physiology and
metabolism (Huang and McDonald, 2009). Facilitating extracellular
secretion of the foreign protein is often an effective strategy in
alleviating this response.
3.1.1.2. Inducible promoters. Regulated or inducible promoters have
been increasingly used in recent years, particularly those which allow
external regulation by chemical stimuli such as alcohol, steroid, salts
and sucrose, or environmental factors such as temperature, light,
oxidative stress and wounding (Boetti et al., 1999; Corrado and
Karali, 2009; Cramer et al., 1996; Huang and McDonald, 2009; Lee
et al., 2006; Padidam, 2003; Tang et al., 2004). Inducible expression is
advantageous as this allows protein production to be separated
from cell growth, thus enabling independent optimization of both
processes. Such regulated expression is especially benecial when the
synthesized proteins are lethal to host cells. In a highly efcient
inducible expression system, the inducer/promoter pair should meet
the following criteria: high specicity, rapid response of promoter
upon induction, rapid switching off promoter upon inducer withdrawal, non-toxic to plant cells and easy scale-up (Sharma and
Sharma, 2009).
The most successful example of an inducible promoter used in
plant cell-based protein expression utilized the rice -amylase 3D
(RAmy3D) promoter which is induced by sucrose starvation. This
promoter has been used for achieving high-level expression in rice
cell suspension culture of many therapeutic proteins including rAAT,
hGM-CSF, hG-CMS, hGH, bryodin-1, lysozyme and human serum
albumin (see Table 1). Virtually every recombinant protein expressed
in this rice suspension cell system has produced signicantly higher
secreted protein levels than attained with any other plant cell
expression system tested. At 247 mg/L, the rAAT protein is the
highest secreted protein recovery reported from plant cell cultures to
date (McDonald et al., 2005); this yield in fact approaches the
production levels delivered by the mammalian cell culture. While

these examples highlight the promise of cell culture systems for


pharmaceutical protein production, the growth rates and characteristics of the rice cell lines are considered inferior to tobacco BY-2 and
NT-1 cell lines (Hellwig et al., 2004), and their viability is negatively
impacted when cultivated in a sucrose-starvation medium (Huang
and McDonald, 2009). Furthermore, requisite medium exchange to
create a sucrose-starvation environment in the cultures presents
signicant technical difculties when considering large-scale production (Park et al., 2010; Su and Lee, 2007).
The oxidative stress-inducible peroxidase (SWPA2) promoter has
also been used for high-yield expression of human lactoferrin (up to
4.3% TSP) in both tobacco BY-2 and Siberian ginseng cell cultures (Choi
et al., 2003; Jo et al., 2006). The SWPA2 promoter has demonstrated
30-fold higher activity than the constitutive CaMV35S promoter in
driving foreign protein expression in plant cells (Lee et al., 2006).
Other chemically inducible promoters used in plant cell cultures
include an estradiol-inducible chimeric XVE transcription activator
(Zuo et al., 2000) and a dexamethasone-inducible pOp/LhG4 transcription activator (Samalova et al., 2005). In addition, plant viralbased expression system that is more efcient for the iterative
replication of viral vector in host cells (Matsuo et al., 2007) have been
developed for inducible expression of foreign genes. Examples include
rAAT expression using the estradiol-inducible cucumber mosaic virus
viral amplicon (CMViva) in tobacco (N. benthamiana) cells (Huang
et al., 2010), GFP expression with a estradiol-inducible tomato Mosaic
Virus (ToMV) amplicon in tobacco BY-2 cells (Dohi et al., 2006) and
Norwalk virus capsid protein expression with an ethanol-inducible
Bean Yellow Dwarf Virus (BeYDV) amplicon in tobacco NT-1 cells
(Zhang and Mason, 2006).

3.1.1.3. Other transcription modulating strategies. In addition to optimization of promoters, transcription activity can be further improved
by engineering enhancers, activators or repressors in locations up- or
downstream of the core promoter. Enhancers, which are DNA sequences
shown to increase gene expression when placed proximal to the
promoter, typically bind activator proteins and thereby facilitate the
binding of RNA polymerase II to the TATA box. For example, while the
CaMV35S promoter containing only 46 bp of 5-sequence (including
TATA box and the CCAAT consensus sequence) is sufcient for accurate
transcription initiation, sequences between 46 and 105 act as an
enhancer that signicantly increase the transcription level of the base
promoter (Lam et al., 1991). Enhancer sequences can also be stacked
to further augment gene transcription. In the case of the CaMV35S
promoter this enhancer sequence was duplicated (or double enhanced;
CaMV35SDE) and shown to increase the expression level of many
heterologous proteins in plants (Kay et al., 1987; Mishra et al., 2006;
Ruggiero et al., 2000; Sharm et al., 2008). Transcription has also been
shown to be enhanced by anking the transgene with sequence
encoding nuclear scaffold/matrix attachment regions (S/MARs) that are
important in mediating structural organization of eukaryotic chromatin (Halweg et al., 2005). In the context of plant transgene expression,
S/MARs have been shown to minimize silencing (Tang and Wang,
2006), increase plant cell transformation frequencies, and reduce
variance of transgene expression (Allen et al., 2000; He et al., 2008;
Petersen et al., 2002). In addition, synthetic super-promoters that
combine the most active sequences of several promoters is another
strategy for boosting gene expression (Sharma and Sharma, 2009). An
example of this approach resulted in a hybrid promoter containing
elements from both the CaMV35S promoter and the Agrobacterium Ti
plasmid mannopine synthetase promoter that increased GUS expression
by 3- to 5-fold over levels achieved with the double enhanced
CaMV35S promoter (Comai et al., 1990; Desai et al., 2010). Although
these strategies have been exploited most extensively in the wholeplant system in boosting heterologous protein expression, they are
generally applicable to plant cell-based systems.

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

3.1.2. Improving translation efciency


The translational efciency of a transgene is largely determined by
the proper mRNA processing (capping, splicing and polyadenylation)
and stability. The 5- and 3-untranslated region (UTR) of the plant
expression cassettes play critical roles in properly processing mRNA and
maintaining mRNA stability (Cowen et al., 2007). While the 5-UTR or
leader sequence is very important for 5-capping and facilitating
translational initiation, the 3-UTR generally is important for transcript
polyadenylation which strongly inuence the stability of mRNA (Abler
and Green, 1996; Chan and Yu, 1998). These untranslated sequences can
be manipulated for the optimization of foreign protein expression
(Sharma and Sharma, 2009). Classic examples include the translational
elements from tobacco etch virus (TEV), tobacco mosaic virus (TMV)
and alfalfa mosaic virus (AMV). These 5-leader sequences used in plant
expression systems enhance transgene expression by several folds by
enabling more efcient translation of the downstream transgene (Datla
et al., 1993; Gallie et al., 1995; Hood et al., 1997; Kang et al., 2004; Wang
et al., 2001). Recently, a cow pea mosaic virus (CPMV) translational
element was developed for extremely high-level and rapid transient
protein expression in plants (up to 25%30% TSP) (Sainsbury et al.,
2008; Sainsbury and Lomonossoff, 2008). Use of this translational
enhancer in N. benthamiana cell culture resulted in an increase in GUS
protein production similar to that obtained with the TEV 5'-UTR
(presented at 2010 Society In vitro Biology meeting; W. Curtis et al.). In
addition to leveraging 5-leader sequence, utilization of a heterologous
3-UTR derived from plants or plant viruses generally stabilizes the
transcript and in turn improves overall protein expression levels
(Cowen et al., 2007; Hood et al., 1997; Staub et al., 2000). In addition,
it has been demonstrated that some AU-rich sequences destabilize
mRNA and removal of this repetitive run from 3-UTR is requisite in
avoiding rapid degradation of the mRNA (De Rocher et al., 1998;
Schillberg et al., 2003).
Another potentially useful approach for improving translational
efciency involves optimization of recombinant gene codon usage to
better match that used by the host production (Gustafsson et al.,
2004; Hershberg and Petrov, 2008). The usage frequency of the
genetic code in plants in fact differs not only from that of animals and
microorganism (Desai et al., 2010), but also between monocots and
dicots (Kawabe and Miyashita, 2003; Liu and Xue, 2005; Murray et al.,
1989). Therefore, using preferred codons and/or removing rare
codons that favor the host plant cell has been shown to be an efcient
means of improving recombinant protein yields (Jabeen et al., 2010).
Among the more striking examples of this codon optimization
strategy was reported for human acetylcholinesterase expression in
tobacco plants which resulted in a 5- to 10-fold increase in
recombinant protein accumulation as compared to expression using
the native human sequence (Geyer et al., 2007).
3.1.3. Minimizing post-translational protein degradation
Evidence of post-translational protein degradation by proteases
within plant cells has been reported in many studies (Callis, 1995;
Doran, 2006a; Schiermeyer et al., 2005; Sharp and Doran, 2001a,
2001b; Stevens et al., 2000; Yang et al., 2005). Loss of synthesized
protein as a result of proteolytic degradation along the secretory
pathway is an important but often overlooked contributing factor to
low protein yields (Doran, 2006a; Shih and Doran, 2009). However,
proteolytic degradation may be effectively minimized by targeting the
foreign proteins to sub-cellular compartments such as the endoplasmic reticulum (ER). Since proteolysis is more likely to occur in the
apoplast and cytosol than the ER, retaining the expressed protein in
ER with the use of an ER retrieval signal (e.g. KDEL, HDEL) has been
commonly used to improve foreign protein stability (Desai et al.,
2010; Doran, 2006a; Sharma and Sharma, 2009). Furthermore, the ER
contains many molecular chaperones that facilitate nascent proteins
folding and assembly (Nuttall et al., 2002) and thus has often been
regarded as an ideal compartment for accumulating many classes of

285

foreign proteins. With ER retrieval, functional protein yields have


been reported to improve by 10- to 100-fold as compared to
recombinant proteins allowed to process through the secretory
pathway uninterrupted (Doran, 2006a; Faye et al., 2005; Hellwig
et al., 2004; Torres et al., 1999; Yasuda et al., 2006). One of the more
dramatic examples was shown for KDEL-targeted human EGF in
tobacco cells where a 104-fold increase in protein yields was
attributed directly to ER retention of the recombinant protein
(Wirth et al., 2004). However, this strategy is not always appropriate,
as is the case with some therapeutic proteins that require signicant
post-translational processing in the Golgi for functionality (Gomord
and Faye, 2004; Kaufman, 1998; Shaaltiel et al., 2007).
Many other strategies have been used for reducing the effects of
proteolytic degradation in plant cells including: co-expression of
recombinant protein with protease inhibitors, co-expression with
protein co-factor and subunit, knockout mutations in the genes
encoding specic proteolytic enzymes, and removal of proteasespecic sites from foreign proteins using genetic engineering
techniques. Reports utilizing co-expression of protease inhibitors
have shown no signicant inhibitory effects on plant growth and
development (Van der Vyver et al., 2003; Xu et al., 2004). Coexpression of recombinant serine proteinase inhibitor II (sPI-II) has
proven successful in increasing hGM-CSF yields in a rice cell
suspensions by 2-fold (Kim et al., 2007; Kim et al., 2008b). Likewise,
co-expression of protease inhibitor (Bowman-Birk Serprotease inhibitor) in transgenic tobacco root cultures resulted in increased
accumulation of a recombinant monoclonal antibody (Komarnytsky
et al., 2006). Protein degradation may be circumvented with some
foreign heteromeric proteins when cofactors and/or subunits are
allowed to be co-expressed (Desai et al., 2010; Doran, 2006a). In the
case of the ricin A-chain when expressed in tobacco cells without its
partner B-chain was easily degraded, while co-expression of both A
and B chains resulted in the formation of disulde-linked heterodimers that were secreted and recovered from the media fraction
(Frigerio et al., 1998). Similarly, co-expression of antibody with its
antigen was reported to increase the antibody yields possibly because
the antibody was stabilized by antigen binding (Stoger et al., 2002b).
While strategies leveraging knockout mutations in the genes encoding select proteinases and removal of protease-specic sites from
target proteins may be effective at reducing post-translational protein
degradation and improving protein stability in plant cell cultures
(Doran, 2006a), few studies in planta have been reported corroborating these approaches.
3.1.4. Stabilizing protein fusions and Hyp-Glyco technology
Expression of a heterologous protein as a peptide- or proteinbased fusion can be used to stabilize the structure of the target protein
in achieving higher protein yields. Utilizing a strategy that incorporates as the fusion partner that is a highly-expressed and stable plant
protein is one such approach (Streateld, 2007). A C-terminal fusion
of a single plant ubiquitin coding unit to -glucuronidase (GUS)
increased recombinant GUS protein levels by 10-fold (Garbarino et al.,
1995). Recently, a new and attractive fusion protein approach, known
as Hydroxyproline-Glycosylation (Hyp-Glyco) technology has been
shown to dramatically increase secreted protein yields from plant cell
culture systems (Kieliszewski and Xu, 2006; Kieliszewski et al., 2006).
This technology involves expressing heterologous proteins as a fusion
with a novel, Hyp-glycomodule tag comprised of Hyp-rich peptides
(HypRP) such as tandem repeats of Ser-Hyp or Ala-Hyp (Fig. 4). These
HypRP tags direct extensive Hyp-O-glycosylation in plant cells
resulting in arabinogalactan polysaccharides populating this repetitive peptide fusion (Kieliszewski, 2001; Shpak et al., 1999; Tan et al.,
2003) and signicantly facilitating the secretion of the expressed
protein from cultured plant cells. Heterologous proteins including
enhanced green uorescence protein (EGFP), human interferon 2
(hIFN2) and hGH have been expressed in tobacco and Arabidopsis

286

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

Fig. 4. Schematics of Hyp-Glyco technology. Expressing therapeutic proteins as fusion with a HypRP tag comprised of tandem Ser-Pro repeats. A typical Hyp-arabinogalactan
polysaccharide (Hyp-glycan) consists of galactose, arabinose, rhamnose and glucuronic acid. The Hyp-glycan structure is from Xu et al. (2007).

cells using the Hyp-Glyco technology. Upwards of 120 mg/L of EGFP,


35 mg/L of hGH and 28 mg/L of hIFN2 were secreted into cell culture
medium, representing a 500- to 1500-fold greater recovery than
their non-glycosylated controls (Shpak et al., 1999; Tan et al., 2003;
Xu et al., 2010, 2005, 2007, 2008; Zhao et al., 2002). Glycosylation of
HypRP tags also greatly extended the serum half-life of small therapeutic proteins including hGH and hIFN2 by as much as 13-fold
without signicantly affecting their respective bioactivity (Gomord
et al., 2010; Xu et al., 2010, 2007). While most therapeutic
applications will require cleavage of this HypRP tag from the fusion
protein and a concomitant increase in downstream processing costs,
this plant-centric Hyp-Glyco technology is extremely promising for
overcoming current limitations linked with low protein yields of
plant-based bioproduction.
3.2. Cell culture approaches
While on average molecular strategies typically increase transgene
expression levels by two to three orders of magnitude (Desai et al.,
2010; Huang and McDonald, 2009), the nal protein productivity of a
plant cell culture system can also be signicantly impacted by the
culture conditions. Factors including media composition, temperature, pH, light and presence of growth regulator or stress factors in
the cultures, are all important determinants of cell growth and
protein production (Rao and Ravishankar, 2002). A high volumetric
productivity is fundamental to pursuing cell culture process development and is crucial for the ultimate commercialization of this
technology. Unlike eld-grown plants, plant cells are cultured in
conned environments, and thus culture conditions can be altered
and manipulated much more readily than in the eld. Consideration
for not only culture conditions but aspects related to retrieval and
recovery of high quality recombinant protein product is an essential
rst step in considering scale-up and process development of plant
cell culture systems.
3.2.1. Higher order testing of culture medium
Although standard plant cell/tissue culture medium such as MS
(Murashige and Skoog, 1962), SH (Schenk and Hildebrandt, 1972),
Gamborg B5 (Gamborg et al., 1968) and LS (Linsmayer and Skoog,
1965), provide sufcient nutrients necessary for plant cell growth,
protein productivity can be further improved by manipulating the
medium composition (Zhong, 2001). The components that have been
routinely adapted include components of the basal medium (nitrogen
and phosphorus macronutrients as well as other micronutrients), the
carbon source (sucrose and glucose) and the phytohormones (auxin
and cytokinin). Since plant cell species often require distinct optimal

conditions for cell growth and foreign protein production, statistical


experimental design approaches are typically employed for customizing the culture medium (Kawana and Sasamoto, 2008; Omar et al.,
2004; Prakash and Srivastava, 2005; Weathers et al., 1997; Xu et al.,
1998b). Holland et al. (2010) reports a 1020-fold increase in the
production of the full-size human anti-HIV antibody in tobacco BY-2
cell cultures through an extensive media analysis and optimization,
which was signicantly impacted by the nitrogen source. In the case
of IgG1 production in tobacco cell culture, medium manipulation has
also been reported to result in a 5-fold increase in yield (Doran, 2000).
In addition, strategies that address potential limitations in the protein
production components such as amino acids can overcome production hurdles; when these protein synthesis precursors were supplemented into the plant cell culture system, a transient 3-fold increase
in antibody levels was observed (Fischer et al., 1999b).
In addition to various nutrients and phytohormones essential to
plant cell growth, other agents are sometimes required for improving
recombinant protein quality and recovery. In particular agents
including bovine serum albumin (BSA), gelatin, polyethylene glycol
(PEG) and polyvinylpyrrolidone (PVP), have been historically used to
stabilize heterologous proteins secreted into the media (Fischer et al.,
1999b; Hellwig et al., 2004; Tsoi and Doran, 2002). While secretion of
a heterologous protein into cultured plant cell medium is potentially
advantageous for downstream processing, these recombinant products can become more susceptible to degradation by proteolytic
enzymes or destabilization due to the simplistic make-up of plant cell
culture medium (Bateman et al., 1997; Doran, 2006a; Huang and
McDonald, 2009; Shih and Doran, 2009). The use of the proteinstabilizing agent PVP (0.75 g/L) in the culture medium resulted in a
35-fold increase of heavy chain (HC) antibody yields compared to that
obtained from control medium lacking PVP (Lee et al., 2000;
Magnuson et al., 1996). Non-proteinaceous agents such as PVP
(LaCount et al., 1997; Shin et al., 2010b; Wongsamuth and Doran,
1997), PEG (Lee et al., 2002), dextran (Lee et al., 2000), Pluronic F-68
(Lee and Kim, 2002), dimethylsulfoxide (DMSO) (Wahl et al., 1995),
as well as proteinaceous agents including gelatin (Kwon et al., 2003b;
Shin et al., 2010b; Wongsamuth and Doran, 1997), BSA (James et al.,
2000) and bacitracin (Bateman et al., 1997) have been utilized in plant
cell culture foreign protein expression. When selecting proteinstabilizing agents, these compounds should be non-phytotoxic at
the concentrations employed and preferably do not interfere with cell
viability, growth and division or the downstream purication process
(Lee et al., 2000).
The precise mechanism by which many such protein-stabilizing
agents function to enhance protein recovery is not clearly understood.
In general these compounds can protect the protein from hydrolytic

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

degradation, prevent protein precipitation, and block nonspecic


interactions between the secreted polypeptide and the glass or plastic
walls of the culture vessel (Doran, 2006b; Hellwig et al., 2004; Lee
et al., 2000). Although protease inhibitors can be added into culture
medium to mitigate protein degradation, these reagents are generally
short-lived in medium and can be cost-prohibitive for large-scale
culture production (Huang and McDonald, 2009). In addition,
exogenously added compounds to the plant cell cultures may also
increase the actual rate of protein synthesis as has been reported for
both gibberellic acid and haemin (Hellwig et al., 2004; Tsoi and Doran,
2002). Indeed signicant benet can be gained by identifying a
culture medium environment early in the production timeline that
addresses the integrity and stability of the specic foreign protein
being produced.
3.2.2. Cell immobilization
The use of cell immobilization has found widespread utility in
plant cell culture. This technique was originally developed to enhance
the production of secondary metabolites, but has proven useful for the
production of heterologous proteins as well (Koyama and Seki, 2004).
Immobilization approaches tested in plant cell cultures include barrier
retention of cells using at membranes and hollow ber membranes
(Jose et al., 1983; Kim et al., 1989; Shuler et al., 1986), encapsulation
or entrapment of cells within a porous matrix such as calcium
alginate, agar and polyurethane foam (Bodeutsch et al., 2001;
Brodelius and Mosbach, 1982; Cappelletti et al., 1999; Gillet et al.,
2000; Liu et al., 1999; Osuna et al., 2008; Ziyad-Mohamed and Scragg,
1990), and surface immobilization of plant cells on the brous
material such as glass bers (Archambault et al., 1989, 1996;
Campostrini et al., 1996). Immobilization has several associated
benets over free cell culture including protection of the cells from
hydrodynamic shear stress, prevention of unintended cell removal
with the efuent in continuous cultures, and convenient recovery of
the cells from culture broth for repeated use without the necessity of
inoculation. It has been shown that cell encapsulation in alginate,
carrageenan or agar could increase the production of hGM-CSF in
tobacco cells by 50% (Bodeutsch et al., 2001), with alginate exhibiting
the highest quality beads and most reproducible growth results. The
mechanism of action is speculated to be most likely due to the
extended exponential growth phase of the cells trapped within the
alginate beads. However, there are limitations with several of these
immobilization techniques currently in use. Issues of both cell leakage
and bead disruption as culture growth progresses have been
associated with porous matrix beads such as calcium alginate.
Likewise, polymeric matrices and barrier membrane systems impose
additional limits on both nutrient and metabolite mass transfer
(Hooker and Lee, 1990). The limitations of immobilization plant cell
culture systems remain a challenge and need to be addressed for this
technology to become commercially viable.
3.2.3. In situ protein removal
Although stabilizing agents have been shown to be effective in
preventing the loss of foreign proteins from attack by proteases,
potential for producing harmful effects on the cells and negatively
impacting downstream processing remains an issue (Joo et al., 2006).
In situ protein removal represents an alternative approach to improve
the recovery of the secreted foreign proteins. This can be achieved
through aqueous two-phase culture (Dutta et al., 1994; Hooker and
Lee, 1990; Ilieva et al., 2001), supplementation of protein-binding
resins (Sharp and Doran, 2001b), and perfusion culture (Lee and Kim,
2006; Su et al., 1996). An aqueous two-phase system (ATPS) is
formulated by mixing two different water-soluble polymers such as
PEG and dextran, or a polymer and a salt such as PEG and phosphate
or sulfate (Cabral, 2007; Ilieva et al., 2001). ATPS which has been used
in a variety of applications including protein extraction (Cabral, 2007;
Hooker and Lee, 1990; Lee et al., 2004a), and may be advantageous for

287

plant cell cultivation since cultured cells are typically immobilized in


one phase along with nutrient substrates while the secreted products
are drawn into and collected from the other phase. This concept of
separate phases may in turn protect the secreted proteins from
degrading proteases associated with the alternate phase and thus
enhance foreign protein productivity. Compared with cell immobilization in/on solid phases, the growth of cells in ATPS is unhindered
since cells remain in a free suspension rather than bound within a
matrix or between barrier membranes (Hooker and Lee, 1990). In
addition, ATPS can achieve high degrees of phase contact which
diminishes limitations associated with mass transfer. Tobacco
suspension cells have been successfully grown in various ATPS
cultures with the highest observed growth rates occurring with a 3%
PEG (20,000)/5% dextran system, wherein the growth rate and cell
density approached that of cultures grown in standard medium
(Hooker and Lee, 1990). Ilieva et al. (2001) reported phosphodiesterases production in tobacco cell culture using an ATPS with 4% PEG
(20,000)/7.5% dextran where the phosphodiesterases partitioned
primarily to the bottom dextran phase and exhibited high specic
enzyme activity.
Addition of resins into culture medium for the binding and
recovery of target metabolites has long been used for enhancing
secondary metabolite production (Bisaria and Panda, 1991; Choi et al.,
2001; Roja et al., 2005). A similar strategy could also be harnessed
for improving recombinant protein recovery which in effect would
isolate the protein product from degrading inuences and/or remove
the protein from product inhibition pathways (James et al., 2002). In
the case of tobacco cells producing and secreting mouse HC mAb and
6xHis tagged hGM-CSF, in situ recovery using Protein G and iminodiacetic acid metal afnity resins, respectively proved quite effective. This approach resulted in as much as an 8-fold increase in HC
mAb production and a 2-fold enhancement of hGM-CSF yield when
compared to these proteins recovered under normal culture procedures (James et al., 2002). In another example, addition of
hydroxyapatite resin to a tobacco NT-1 cell culture with periodic
harvesting of the resin led to reduced susceptibility of the secreted
IgG1 antibody to hydrolytic degradation and an overall increase in
product recovered (Sharp and Doran, 2001b). Perfusion culture for
in situ removal of protein is typically carried out in a bioreactor, as will
be discussed in further detail in following section. However demonstrating proof-of-principle of this technology, a semi-continuous
perfusion culture of tobacco NT-1 cells conducted in shake ask
performed by replacing 33% of the medium every 12 h, allowed constant
levels of secreted HC mAb to be harvested (LaCount et al., 1997).
3.3. Bioreactor engineering approaches
Process development is a critical step in scaling-up plant cell
cultures to achieve commercial productivity. While similar to
bacterial fermentation or mammalian cell culture in that plant cells
can be grown in bioreactors as homogeneous suspensions, there are
some notable differences in terms of growth kinetics, obtainable cell
densities and nutritional requirements. Although plant cells are
readily cultured in most conventional bioreactors with minor
adjustments (Hellwig et al., 2004; Su and Lee, 2007), scaling-up the
plant cell culture from shake asks to bioreactors is not always
straightforward. Due to the dramatic change of cell growth environments between the two platforms in terms of hydrodynamic shear
forces and rheological properties (Curtis and Emery, 1993; Kieran
et al., 1997; Sajc et al., 2000), poor growth rates of cells and relatively
low product yields are common challenges (Sajc et al., 2000; Su and
Lee, 2007). Fortunately, most of these limitations can be addressed
with improved bioreactor design and optimizing key parameters of
the system including agitation speed, aeration rate and nutrient
supply. Other aspects include identifying plant species most amenable
to in vitro culturing and effective screening procedures for selecting

288

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

Table 2
Enhanced recombinant protein production in plant cell cultures with process engineering approaches.
Culture mode

Bioreactor type and strategies

Expressed protein

Host
plant cell

Yield or productivity enhancement

Reference

Batch
culture

2 L Stirred tank with a single pitched blade impeller.


Control higher medium pH

1-antitrypsin
(rAAT)

N. benthamiana

Huang et al.
(2009)

Fed-batch
culture

7 L and 15 L stirred tank with four-bladed hollow-paddle


impeller. 10 concentrated amino acids fed from day 7.

4.5 mg/L total and 136 ug/L functional


protein, up to 4-fold increase in
productivity
76.5 mg/L or 5.8 mg/L/day, 1.2-fold
increase compared to that of
two-stage cultures with medium
exchange
2-fold increase in volumetric productivity
compared with batch culture
Maximum 40 ug/L/day, 9.4-fold increase
in productivity compared with batch
culture

Human cytotoxic
Tlymphocyte antigen
4-immunoglobulin
(hCTLA4Ig)
Fed-batch
3 L stirred-tank bioreactor. 10 B5 medium with 300 g/L
Green uorescence
culture
of sucrose fed based on culture GFP uorescence
protein (GFP)
Perfusion
5 L (2 L working volume) stirred tank with 4-bladed
Human granulocyteculture
hollowed-paddle impeller. A 50 m stainless steel
macrophage colony
meshes as internal cell separator. Perfusion rates at
stimulating factor
-1
0.5, 1.0 and 2.0 day .
(hGM-CSF)
Perfusion
3.3 L (working volume) stirred tank with a 6-bladed
Acid phosphatase
culture
Rushton turbine on top and a 3-bladed upward pumping (APase)
marine axial impeller on the bottom. A cylindrical bafe
as internal cell separator. Perfusion rate up to 0.4 day 1.
Semi-continuous 2 L (working volume) stirred tank with a single pitched
1-antitrypsin
culture
blade impeller. Medium exchange daily at dilution rates of (rAAT)
1
0.05, 0.1, 0.15, 0.175, and 0.2 day .
1-antitrypsin
Semi-continuous 5 L (4.5 L working volume) stirred tank with a single
culture
pitched blade impeller. Six complete medium exchanges (rAAT)
(three inductions and three additions of fresh growth
medium) for each run lasting ~ 30 days.
Continuous
10 L (9.6 L working volume) stirred tank with two
Carrot acidic
culture
six bladed stirrer turbines. Dilution rate at 0.25 day 1
invertase
1
for 22 day, then 0.2 day
afterwards.

elite cell lines exhibiting satisfactory growth characteristics and


genetic stability (Parekh et al., 2008). In addressing and optimizing
operations, plant cell cultures in bioreactors producing hGM-CSF or
hCTLA4Ig have in fact shown signicantly better performance and
productivity than their shake ask controls (Lee et al., 2004b; Park
et al., 2010). Foreign protein production can be further improved by
applying advanced culture strategies such as fed-batch culture,
perfusion culture, continuous culture and various modications of
these platforms as outlined in Table 2 (Georgiev et al., 2009; Roberts
and Shuler, 1997; Su and Lee, 2007). Bioreactor engineering aspects of
plant cell cultures including bioreactor design criteria and optimization strategies have been recently reviewed (Georgiev et al., 2009;
Huang and McDonald, 2009; Parekh et al., 2008). Thus this section will
summarize some advanced culture strategies in the context of
enhancing foreign protein production with a particular focus on the
use of disposal bioreactors for plant cell cultures.
3.3.1. Optimized bioreactor design and operation
The goal in bioreactor design and operation is to provide a
prolonged and sterile culture environment with efcient mixing and
oxygen exchange in an effort to support optimized cell growth and
achieve high productivity (Su and Lee, 2007). Large-scale culturing of
plant cells has been conducted in variety of bioreactors including

Rice (O. sativa L.)

N. tabacum L. cv
Xanthi
N. tabacum L. cv
Havana SR

Anchusa
ofcinalis

~ 49 units/L/day, more than 3-fold


increase in productivity compared
with batch culture

Park et al.
(2010)

Liu et al.
(2001)
Lee et al.
(2004b)

Su and
Arias
(2003)

Up to 0.6 mg/L/day for individual


cycle, 25-fold increase in productivity
compared with batch culture
Rice (O. sativa L.) 312 mg/L/day for individual cycle;
overall productivities up to
7.7 mg/L/day.

Huang et al.
(2010)

N. tabacum
BY-2

Des Molles
et al. (1999)

N. benthamiana

500 unit/L/day,4-fold increase in


productivity compared with batch
culture

Trexler et al.
(2005)

standard stirred-tank bioreactor (STR) (Doran, 1999; Hooker et al.,


1990), bubble column bioreactor (BCB) (Kim et al., 2001; Min et al.,
2007), air-life bioreactor (ALB) (Hsiao et al., 1999; Su et al., 1996) and
other novel bioreactor types including the wave bioreactor (Eibl and
Eibl, 2006; Terrier et al., 2007), the membrane bioreactor (McDonald
et al., 2005), the hollow ber bioreactor (Kim et al., 1989) and the
rotating drum reactor (Shibasaki et al., 1992; Tanaka et al., 1983).
While lessons can be learned from scaled production of microbial
and mammalian cells, plant cells have many unique and distinguishing features that impose limitations on their large-scale growth
and process development (Table 3). Distinct properties of plant cell
cultures that necessitate special consideration in bioreactor process
development include: large cell size and complex morphology, tendency for aggregation, time-dependent rheological behavior, foaming and wall growth, shear sensitivity, and relatively low growth
and oxygen uptake rate (Parekh et al., 2008). The implications of
these factors on bioreactor design and operation have been comprehensively reviewed (Huang and McDonald, 2009; Sajc et al., 2000).
Some plant-centric factors of mention include the inherent
sensitivity of suspended plant cells to shear forces due to the majority
of their cell volume (~ 90%) being associated with intracellular
vacuoles and the rigid nature of their cellulose-based cell wall
(Dunlop et al., 1994). The shear-sensitivity of plant cells has been

Table 3
Comparison of the characteristics among plants cells, microbes and mammalian cells in bioprocess development.
Characteristics

Plant cells

Mammalian cells

Bacteria

Shape
Size
Cell wall
Doubling time
Cell aggregation or clumping
Shear sensitivity
Oxygen uptake rate (OUR)
KL required in bioreactor operation
Protein yields
Protein localization
Scale-up capacity

Spherical/cylindrical
20200 m
Yes
20100 h
Aggregated to form cell clusters over 2 mm
High
510 mmol/L h
1050/h
0.01200 mg/L
Intracellular/secreted
Medium

Spherical
1050 m
No
2048 h
Not aggregated
Extremely high
0.055 mmol/L h
0.2510/h
110 g/L
Secreted
Medium

Spherical
b1 m
Yes
Less than 2 h
Not aggregated
Low
1090 mmol/L h
1001000/h
b1 g/L
Usually intracellular
High

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

the subject of extensive investigation (Camacho et al., 2000; Namdev


and Dunlop, 1995; Sowana et al., 2001; Sun and Linden, 1999; Zhong
et al., 2009, 1994) however, the precise mechanism by which
shear stress induces cell damage is not well-understood and has
been very difcult to study due to the incredible diversity of plant cell
lines, the varying aggregate size distribution and their assorted cell
morphologies (Huang and McDonald, 2009). As such, susceptibility of
plant cells to shearing forces varies widely among different cell
lines. Even within the same culture line, the age of the culture and the
cultivation schedule can signicantly inuence the responses of the
cells to shear elds (Georgiev et al., 2009). The concept of critical
shear stress, above which cell viability is lost, has been an important
factor in establishing guidance for plant cell bioreactor design (Huang
and McDonald, 2009). The general solution to the shear-sensitivity
issue involves cultivating plant cells under low-shear stress environments such as those established in pneumatic bioreactors (air-lift and
bubble column bioreactor), centrifugal impeller bioreactors (CIB)
(Wang and Zhong, 1996a, 1996b), or stirred-tank bioreactors (STR)
with decreased impeller agitation speed or with a specially designed
low-shear impeller. In the case of STRs others have suggested that
shear sensitivity may not be as critical, and that signicant losses in
viability are not, in general, to be expected under normal operating
conditions in a conventional STR (Kieran et al., 1997).
3.3.2. Advanced bioreactor culture strategies
Batch culture is the simplest culture strategy and is most
frequently employed for large-scale plant cell cultures. Optimizing
culture conditions such as agitation speed (in STR), aeration rate, and
pH, has led to higher protein yields than those obtained from shake
asks (Lee et al., 2004b). However, due to the inherent limitations
of the batch culture mode, the protein productivity of plant cells in
this basic system cannot be further improved. The long lag phase,
the depletion of the key nutrients and the accumulation of inhibitory
substances/metabolites prevent the batch culture from achieving
desired productivity. In addition, signicant amounts of time are
required for sterilizing the culture system hardware and media as well
as lling, emptying and cleaning the system (Georgiev et al., 2009).
This time and labor-intensive issue has been the major driver in the
development of advanced culture strategies, including fed-batch
(Park et al., 2010), two-stage (Shin et al., 2003), perfusion (Wang
et al., 2010), semi-continuous (Su and Arias, 2003) and continuous
cultures (van Gulik et al., 2001) for improving the overall productivity
of plant cell culture technology (Georgiev et al., 2009; Roberts and
Shuler, 1997; Su and Lee, 2007). The integration of these advanced
culture systems for plant cells has been much more limited with
the majority of these linked to secondary metabolite production as
opposed to recombinant proteins. In addition, the choice of culture
strategy can be signicantly impacted by species-specic characteristics of the host cell (specic growth rate, aggregation, etc.), the
promoter used for driving the expression of the recombinant protein
(constitutive or inducible), and the localization and structural/
functional properties of the expressed proteins (intracellular or
secreted; stable or easily degradable; toxic or harmless to hosts)
(Huang and McDonald, 2009).
3.3.2.1. Fed-batch culture. Fed-batch cultures are characterized by the
addition of one or more nutrients continuously or intermittently
into the medium after the start of cultivation, or at some point during
the batch process (Georgiev et al., 2009). Simple implementation
makes this the most popular culture strategy for achieving high
cell density and prolonging the exponential growth phase (Sim
and Chang, 1999). Fed-batch is especially appropriate for growthassociated production systems such as those driven by a constitutive
promoter. Such a medium-feeding strategy demonstrated that
improved cell biomass and protein (GFP) production could be
achieved in a fed-batch culture of tobacco BY-2 cells (Liu et al.,

289

2001). Although fewer studies have been reported, fed-batch culture


has also been shown to improve productivity in an inducible
expression system. In the production of hCTLA4Ig expressed under
the inducible RAmy3D promoter, a fed-batch addition of concentrated
amino acids into the rice cell culture grown in the 15-L stirred-tank
bioreactor increased productivity of the foreign protein by 1.2-fold in
comparison to the more complex two-stage culture that implemented
a medium exchange (Park et al., 2010). However, like batch culture,
protein productivity in fed-batch cultures is also limited by accumulation of toxic or inhibitory metabolites and cell growth has been
shown to be retarded at higher cell densities (above 60% packed cell
volume (PCV)) (Su, 1995).
3.3.2.2. Perfusion culture. Perfusion cultures involve maintaining cells
within a bioreactor where fresh medium is continuously fed and the
spent, cell-free medium is constantly being removed. In comparison
to fed-batch cultures, perfusion culture offers additional process
exibility and advantages. In addition to growth inhibition caused by
the accumulation of inhibitory metabolites in the medium being
alleviated in perfusion culture, the cultured cells in a bioreactor can be
reused thereby minimizing turnaround time between batches and
thus a concomitant increase in the overall productivity of the system.
Perfusion culture has been exploited for the culture of plant cells
with notable success (De Dobbeleer et al., 2006; Su and Arias, 2003; Su
et al., 1996; Wang and Qi, 2009; Wang et al., 2010). Cell densities
of 31 g/L were obtained for perfusion cultures of Thalictrum rugosum
cell in a STR (Kim et al., 1991). A major challenge in establishing a
plant cell perfusion culture is maintaining healthy, robust and fully
viable cultured cells in the bioreactor while the cell-free spent
medium is continuously being removed and the fresh medium
supplemented (Su et al., 1996).
The most common approaches used for cell retention in perfusion
cultures include continuous centrifugation (Johnson et al., 1996),
in situ ltration (Kawahara et al., 1994) and gravitational sedimentation (Su and Arias, 2003). Because plant cells tend to grow as
aggregates, cell/medium separation can be accomplished with
relative ease using gravitational sedimentation. Development of an
efcient plant cell perfusion process by inserting a cylindrical bafe
into the STR vessel to create an annular settling zone wherein continuous cell/medium separation via gravitational sedimentation has
been achieved (Su and Arias, 2003). In this study, perfusion cultures
could be maintained at perfusion rates of 0.4 day 1 and a PCV of
60% while successfully achieving complete plant cell retention. In
order to establish a higher rate perfusion culture, (De Dobbeleer et al.,
2006) modied a STR tank for plant cell production by installing four
sedimentation columns vertically on the lid of the tank, which
enabled a maximum perfusion rate of 20.4 day 1 to be run from day 4
to day 6, and a reduced rate of 5 day 1 to be run on day 9 at a 55% PCV.
However, operation of the perfusion culture under extremely high
PCV (e.g. PCV N60%) for a prolonged period will lead to declined
oxygen uptake and reduced cell viability due to the agglomeration
and sedimentation of the plant cells (Huang et al., 2010). In this case,
continuously removing a certain percentage of cultured cells with
the medium in a process referred to as cell bleeding helps maintain
the best performance of the perfusion culture. Cell bleeding at
0.11 vvd (reactor volume per day) had been shown to stabilize the
perfusion culture of Anchusa ofcinalis cells maintained at a PCV below
60% and successfully used for the production of an acid phosphatase
(Su and Arias, 2003). In this study, both the specic oxygen uptake
rate and cell viability were shown to increase, leading to higher cell
biomass yields approaching 25 g/L and improved acid phosphatase
productivity of ~490 units/L/day. Although further optimization of
the culture operation is required, perfusion bioreactor systems are
emerging as the preferred advanced culture strategy relative to other
systems for achieving high productivity of foreign protein expressed
in plant cell culture.

290

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

3.3.2.3. Continuous culture/semi-continuous culture. In a continuous or


chemostat operation, fresh medium is continuously fed into the
bioreactor and the excess culture (spent medium and cells) is
simultaneously washed out, thereby permitting the maintenance of
cultures very close to their maximum growth rate. Due to long runtimes and the need for plant cells to be maintained in exponential
growth phase, continuous culture is attractive and holds promise as
an emerging advanced culture strategy (Georgiev et al., 2009).
However the limited research that exists on this continuous culture
system has primarily focused on secondary metabolite production
(Miller et al., 1968; Sahai and Shuler, 1984; van Gulik et al., 2001).
Some typical problems associated with this culture strategy include
low specic growth rate, cell aggregation, genetic instability and
excretion of sticky polysaccharides (Hellwig et al., 2004; van Gulik
et al., 2001). These problems make it difcult to maintain an ideallymixed suspension of cells which is requisite for establishing a
continuous culture system.
A modication of this system known as semi-continuous culture
is a hybrid of batch and continuous operations and may be a more
appropriate application for plant cell-based recombinant protein production. In a semi-continuous operation, a portion of the suspension
culture (spent medium and cells) is periodically harvested in exchange
for an equivalent volume of fresh medium. The advantages of such
a semi-continuous culture include: active recombinant proteins can
be harvested periodically and factors limiting cell growth and protein
stability in the culture broth are simultaneously removed (Huang
et al., 2010). Long-term chloramphenicol acetyltransferase (CAT)
production has been demonstrated with a 41-day, semi-continuous
culture of tobacco cells in a STR bioreactor (Hogue et al., 1990). Semicontinuous cultures may also be suitable for inducible production
system, where a two-stage culture is often practiced; rst stage
dedicated to cell growth and the second to protein production. An
example of this conguration was shown with a rice cell culture
system expressing a transgene under the inducible Ramy3D promoter
(Huang and McDonald, 2009). In this case an exchange to sugar-free
medium was required at a specic phase of cell growth to induce
the recombinant protein expression. The cyclical semi-continuous
operation enabled culture conditions to be alternated to support
either cell growth or production as well as the reuse of the cultured
rice cells in promoting a long-term operation. With a semi-continuous
culture of rice cells consisting of three cycles over a 2528 day period,
rAAT protein levels ranging from 4.5 to 7.7 mg/L/day were achieved
(Trexler et al., 2005). More recently, Huang et al. (2010) developed a
semi-continuous bioreactor operation for enhancing the production
of rAAT in N. benthamiana cell culture. In this optimized semicontinuous operation, plant cells transformed with the inducible
CMV viral amplicon and expressing rAAT maintained higher viability
and produced lower extracellular protease activity and phenolic
concentrations than when grown in a batch mode. Integration of
this semi-continuous culture method resulted in a 25-fold increase in
the production of rAAT that corresponded to 603 g/L of protein
harvested daily.
3.3.3. Adoption of disposable bioreactors
In recent years there has been a trend towards the use of
disposable bioreactors for large-scale plant cell culture with a goal
of reducing production costs and minimizing validation efforts under
GMP regulations (Ducos et al., 2010; Eibl et al., 2010; Hacker et al.,
2009). In contrast to traditional glass or stainless steel bioreactors,
disposable bioreactors are usually made of FDA-approved biocompatible plastics such as polyethylene, polystyrene, polytetrauorethylene, polypropylene and ethylene vinyl acetate (Eibl and Eibl, 2006;
Eibl et al., 2010). Since the bioreactor vessel is pre-sterilized and
discarded after harvest, clean-up and re-sterilization steps are
eliminated and associated risks with culture contamination are
reduced (Singh, 1999). To date the integration of disposable

bioreactors for plant cell culture and particularly for the production
of foreign proteins is surprisingly limited. However one example that
highlights the integration of this type of advanced culture technology
is the commercial production of the human recombinant glucocerebrosidase in carrot cells grown in exible plastic bags (www.protalix.
com). Inspired by the historical success of Protalix, adoption of
disposable bioreactors for plant cell culture at large-scale is
anticipated to increase due to the clear advantages of process
simplicity, safety and production exibility.
The various types of disposable bioreactors currently used for plant
cell culture include wave-mixed, orbitally shaken or stirred reactors
and have been recently reviewed (Eibl et al., 2010). Schematics and
applications of the major types of disposable reactors that have been
used for plant cell culture are listed in Table 4. The wave bioreactor
represents the most successfully developed type of disposable
bioreactor to date. Introduced about 10 years ago (Singh, 1999),
units with working volumes up to 300 L (Eibl and Eibl, 2008) have
been commercialized by Wave Biotech LLC, now part of GE Healthcare.
In the wave bioreactor operation, cells are grown in an inated bag
placed on a rocking platform that induces wave action of the liquid
medium (Singh, 1999). Although originally developed for mammalian
cell culture, the wave bioreactor has successfully been used for
culturing a range of plant suspension cell types sourced from tobacco,
grape, apple, yew and barley at culture volumes ranging from 0.4
to 10 L and with biomass productivity approaching 40 g(FW)/L/day
(Eibl and Eibl, 2008; Ritala et al., 2008). However, the volumetric
oxygen transfer efciency (kLa) of the wave bioreactor is typically low
(b4 h 1) (Singh, 1999), resulting in possible oxygen limitation with
high cell-density cultures.
In contrast to the wave bioreactor that rocks the whole bag, the
wave and undertow (WU) bioreactor moves only one section of the
bag up and down to form waves in the bag. This has resulted in
improved kLa values approaching 10 h 1 in 30 L WU bioreactor
cultures of tobacco BY-2 cells (Terrier et al., 2007). The slug bubble
(SB) bioreactor is made up of a vertical plastic cylinder lled up to
80% of its height with the cultured cell suspension (Ducos et al., 2010).
Single slug bubbles are generated at the bottom and rise up to the top
of the cylinder thereby providing agitation and aeration. Much higher
kLa values approaching 17 h 1 for a 50 L SB bioreactor culture of BY-2
cells have been obtained in this system (Terrier et al., 2007). The
plastic-lined bioreactor was rst reported by Hsiao et al. (1999) and
later patented for growing Hyoscyamus muticus cells (Curtis, 2004).
This type of bioreactor is constructed by inserting a sterilized plastic
liner into a vessel attached with a head plate. Aeration and mixing are
provided by an aeration tube passing through the head plate down to
the bottom of the cell culture. Similar to the plastic-lined bioreactor,
the stirred bag bioreactor consists of a plastic bag in a steel support
container equipped with aeration devices and rotating or bumbling
impellers. The suitability of a 50 L stirred bag bioreactor for growing
tobacco cells was demonstrated by Eibl et al. (2009). Finally, the
Osmotek bag bioreactor is made up of a simple culture bag without
any exterior supporting vessel. This system was developed originally
by Osmotek LTD (Rehovot, Israel) to grow tissues and more recently
has been modied by Protalix to accommodate large-scale plant cell
cultures used in producing therapeutic proteins (e.g. glucocerebrosidase) by employing an external open wire cage around the bag
(Shaaltiel et al., 2007; Weathers et al., 2010). This system in fact will
be the bioreactor used in producing the rst plant-made, human
therapeutic to be FDA-approved and successfully commercialized
(Desai et al., 2010).
3.4. Downstream processing
Although high productivity is critical in moving plant cell culture
technology towards commercialization, the efcient recovery and
purication of target recombinant proteins from either cultured cells

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

291

Table 4
Disposable bioreactors used for plant cell cultures.
Reactor type

Cultured cells

Products

Working volume

Yields/productivity

Reference

Wave bioreactor

Vitis vinifera
N. tabacum BY-2
Hordeum vulgare L.

Biomass
Biomass
Humancollagen I
alpha1

1L
10 L
4L

40 g(FW)/(L day)
32 g(FW)/(L day)
5.1 g/L (day 25)

Eibl and Eibl (2006)


Eibl and Eibl (2006)
Ritala et al. (2008)

N. tabacum BY-2

Biomass

10 L
20 L
30 L
100 L

13.6 g(DW)/L
12.8 g(DW)/L
12.6 g(DW)/L
13.0 g(DW)/L

Terrier et al. (2007)

N. tabacum BY-2

Biomass

10 L
20 L
50 L
70 L

17.2 g(DW)/L
13.7 g(DW)/L
14.2 g(DW)/L
12.9 g(DW)/L

Terrier et al. (2007)

Plastic-lined bioreactor
G
A
S

Hyoscyamus muticus

Biomass

28.5 L
100 L

7.02 g(DW)/L (day 13)


15.0 g(DW)/L (day 33)

Hsiao et al. (1999)


Curtis (2004)

Stirred bag bioreactor

N. tabacum BY-2

Biomass

25 L

13.5 g(DW)/L (day 5)

Eibl et al. (2009)

Carrot cells

Glucocerebrosidase

N/A

N/A

(Shaaltiel et al., 2007;


Weathers et al., 2010)

Wave and undertow bioreactor

Slug bubble bioreactor


G

Slug Bubble

AG

Osmotekbag bioreactor
G

Aair inlet; Gexhaust gas; Ssampling; DWdry weight; FWfresh weight; N/Anot available.

or extracellular medium is equally important. As in other cell-based


bioproduction systems, the downstream processing in plant cell
culture may account for as much as 80% of the total production costs
(Hellwig et al., 2004). The purication of proteins for therapeutic
application represents the most challenging and expensive process
in which protein of the highest purity (N99%) is required and purication procedures must meet the stringent regulatory standards
established that govern production of biopharmaceuticals (Hellwig
et al., 2004). Therefore, efforts to develop new and improved downstream processing methods can signicantly reduce the overall costs
associated with plant cell-based production systems (Nikolov and
Woodard, 2004).
In plant cell expression systems, the target proteins are either
retained inside the cells or secreted into the medium. Secretion
systems are advantageous to the downstream protein recovery
process in that typically two unit operation steps cell disruption
and clarication can be circumvented (Fig. 5). Generally lower in
phenolic compounds and other oxidizing substances, the medium
fraction enters the purication process containing a much lower
percentage of contaminating proteins and other metabolites relative
to cell extracts (Hellwig et al., 2004). On the other hand secreted
target proteins may be diluted and thus more unstable in the culture

Fig. 5. A standardized procedure of protein separation and purication from plant cells
or cell culture medium.

292

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

medium highlighting the need for efcient and rapid recovery


techniques for the foreign protein from large medium volumes. In
contrast, the advantages of intracellular retention for downstream
processing lie in the smaller volume of the harvested plant cells and
in some cases a higher concentration of the target protein in the
starting material. Major disadvantages associated with cell retention
of the foreign protein include a more complex composition of the
feed-stream and the liberation of proteolytic and oxidizing compounds during the cell disruption process (Hellwig et al., 2004).
Various considerations of downstream processing for transgenic
plant-derived proteins have been previously reviewed (Hellwig
et al., 2004; Menkhaus et al., 2004).
The downstream processing for plant cells where the protein is
intracellularly retained begins with cell disruption/clarication, and is
followed by concentration/capturing and chromatographic purications (Fig. 5). Some examples of downstream processing studies with
plant cell culture are shown in Table 5. While secreted proteins can
advance immediately to the concentration/capturing step of the
process, those retained inside cells must rst be released by breaking
the cell wall and membrane. Techniques available for plant cell
disruption include sonication, pressure homogenization, wet milling
and enzymatic lysis. While all methods work efciently in breaking
the plant cell wall, wet milling and sonication are in fact difcult to
scale-up and enzymatic lysis is cost-prohibitive at scale (Hellwig et al.,
2004). Thus, pressure homogenization is generally regarded as the
method of choice in large-scale processes. After cell disruption,
clarication of the cell extract is typically carried out by either deadend or cross-ow ltration for large-scale production or centrifugation at smaller scale. Alternatively, advanced chromatographic
technology Expanded Bed Adsorption (EBA) could be an efcient
approach for simultaneous clarication, concentration, and prepurication from either cell extract or cell culture medium (Bai and
Glatz, 2003; Valdes et al., 2003).
The purication steps that follow are generally common to the
protein recovery methods used in other production platforms. Several
chromatography options including ion exchange chromatography
(IEX), afnity chromatography (AC), hydrophobic interaction chromatography (HIC) and size-exclusion chromatography (SEC) are used
for the enrichment of the recombinant protein (Fig. 5). Method
development from selection of the most effective chromatographic
matrices, to their arrangement in a suitable order and evaluation of
their efciency in product recovery is dependent upon the unique

properties of the target protein including molecular size, charge,


solubility, and stability as well as the protein background of the
production host. The use of afnity tags including GST (glutathione
S-transferases), MBP (maltose binding protein) and 6xHis can facilitate
the capture of the foreign protein but these tags may require removal
following purication to restore the native structure/function of the
protein or is required for market distribution of the product (Fischer
et al., 2004). In the case of recombinant antibody production in plants
(Plantibodies), Protein A or Protein G afnity chromatography provide
efcient and convenient choices for protein capture and purication
(Girard et al., 2006; Hellwig et al., 2004; Hussack et al., 2010; Park et al.,
2010). In addition to the column-based separation approaches
mentioned above, selection based on membranes such as ultraltration, microltration, pervaporation, and pertraction should also be
considered for use in protein purication. The advantages of membranes
include speed, easier scale-up and large interfacial area available for
mass transfer (Charcosset, 2006; Sajc et al., 2000). Ultraltration has
recently been used to purify the recombinant type I human collagen and
GFP from corn extracts with success (Aspelund and Glatz, 2010).
4. Other challenges to overcome
Outside of low productivity that could be addressed and improved
by implementing the strategies discussed above, other outstanding
challenges for recombinant protein production in plant cell culture
that remain to be addressed include non-mammalian glycosylation
and genetic instability (Doran, 2000; Gomord et al., 2005; James and
Lee, 2006; Shih and Doran, 2009).
4.1. Non-mammalian glycosylation
There are some distinct and important differences in the glycosylation processes of animal and plant cells that warrant consideration when producing recombinant proteins. Although the resulting
alteration of the plant glycosylation pattern may not affect the activity
of the recombinant protein, possible immunogenicity issues in using
plant-produced glycoproteins for animal/mammalian applications
need to be considered (Doran, 2000; Faye et al., 2005; Garcia-Casado
et al., 1996; Li and d'Anjou, 2009). As the issue of protein N- and Oglycosylation in plants and the limitations and advantages of plantspecic glycosylation on recombinant therapeutic proteins has been
reviewed in depth (Gomord et al., 2010), the following discussion

Table 5
Examples of downstream processing studies with plant cell culture system.
Cell culture system

Expressed protein

Localization of protein

Separation/purication procedure

Purity achieved

Reference

N. tabacum BY-2

Secreted

Xu et al. (2007)

Secreted

N95%

Terashima et al. (1999)

N95%

Huang et al. (2005)

Carrot cell

Glucocerebrosidase

Intracellular retention

N99%

Shaaltiel et al. (2007)

N. tabacum cv. Xanthi

GFP and hGM-CSF-GFP

Intracellular retention

N80%

Peckham et al. (2006)

N. tabacum cv. Xanthi

anti-rabies virus mAb

Intracellular retention

Capturing with HIC;


Purication and polishing with SEC and RP-HPLC
One-step capturing and purication with AC
(with anti-AAT antibody)
Clarication with ammonium sulfate precipitation
and centrifugation;
Capturing and purication with anion IEX
Concentration with membrane ltration
Cell disruption with homogenization;
Clarication with centrifugation;
Capturing with cation IEX;
Purication and polishing with HIC and cation IEX
Cell disruption with ultra-sonication;
Clarication with ammonium sulfate precipitation
and centrifugation;
Capturing with HIC;
Purication with anion IEX
Cell disruption with French press;
Clarication with centrifugation;
Concentration with membrane cross-ltration;
One-step capturing and purication with protein
G-based AC

N99%

O. sativa L. cv Tainung 67

Human interferon 2
(hIFN 2)
Human 1-antitrypsin
(rAAT)
Human serum albumin

N95%

Girard et al. (2006)

O. sativa L. cv Taipei 309

Secreted

IEXion exchange chromatography; ACafnity chromatography; HIC hydrophobic interaction chromatography; SECsize-exclusion chromatography.

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

summarizes this complex topic and specically highlights the issue of


glycosylation as it relates to foreign protein expression in the plant
cell culture platform.
The N-linked glycans produced in plants or plant cells differ from
their mammalian counterparts in the processing and modications of
the core glycan that occur in the Golgi apparatus (Faye et al., 2005;
Frey et al., 2009). There are two key modications on core glycan in
the plant Golgi generally considered the major obstacles limiting the
use of plant-made biopharmaceuticals (Frey et al., 2009): 1) the
absence of (1,4)-galactose, core (1,6)-fucose and terminal sialic
acids that characterize animal glycosylation and which may reduce
the clinical efciency of the plant-derived glycoproteins due to their
decreased serum half-life (Gomord et al., 2010; Lerouge et al., 1998)
and; 2) incorporation of (1,3)-fucose and (1,2)-xylose residues,
two epitopes not found in mammalian glycans that have been linked
with inducing immunogenicity (Bardor et al., 2003; Doran, 2000;
Wilson et al., 1998). Thus far, considerable progress has been made
towards the humanization of protein N-glycosylation in plant cells.
Strategies showing much promise include targeted expression of
therapeutic proteins retrieved from the early Golgi back into the ER
(Lai et al., 2010; Petruccelli et al., 2006), knockout of certain Golgi
enzyme glycosyltransferases (e.g., for xylose and fucose) (Cox et al.,
2006; Koprivova et al., 2004; Sourrouille et al., 2008; Strasser et al.,
2008), utilization of the N-glycosylation plant mutants (Downing
et al., 2006; Strasser et al., 2004) and the engineering of the mammalian glycosyltransferases (e.g. (1,4)-galactosyltransferase or
(1,4)-N-acetylglucosaminyl transferase III) (Bakker et al., 2001;
Frey et al., 2009; Gomez and Chrispeels, 1994). In addition, BecerraArteaga and Shuler (2007) report that culture medium supplementation with nucleotide-sugar precursors such as glucosamine can
alter signicantly the N-linked glycosylation of a recombinant protein
in tobacco cell culture. Most recently, Shin et al. (2010a) report that
sugar starvation conditions, e.g. growth medium with sucrose (+S) or
under sucrose starvation (S), also alter the N-glycan patterns of
the glycoproteins secreted from rice cell suspension cultures. These
studies provide another alternative strategy for the humanization of
N-glycosylated, plant-expressed recombinant proteins.
While N-glycosylation has been extensively addressed, very little
attention has been given to the O-glycosylation of plant cell-made
foreign proteins (Gomord et al., 2010). In general, O-glycosylation is
one of the most complexly-regulated, post-translational modications of proteins, and yet is one of the least understood processes.
Unlike N-glycosylation which occurs at a consensus sequence (Asp-XSer/Thr), there is no well-dened sequence for predicting protein Oglycosylation. In plant cells, O-glycosylation has been described
mainly for the hydroxyl groups of hydroxyproline (Hyp) Ser and
Thr residues, of which the O-glycosylation on Hyp residues is unique
to higher plants and green algae (Kieliszewski and Shpak, 2001;
Showalter, 1993). The Hyp-O-linked glycans are extremely abundant
in plant cells and in fact make a signicant contribution to the
structural properties of the plant extracellular matrix as distinct posttranslational modications of both the extensins and arabinogalactan
proteins (AGPs) (Cassab, 1998; Kieliszewski and Lamport, 1994).
Therefore, in the case of therapeutic proteins produced in plant cells,
the issue of these non-human Hyp-O-glycans being a source of
immunogenicity has been addressed in several studies (Leonard et al.,
2005; Willats et al., 1998; Yates et al., 1996). However to date, all
studies (Xu et al., 2010, 2007) have indicated that the Hyp-O-glycans
produced in tobacco BY-2 cells does not elevate the mouse serum
antibody titers beyond those marginal titers determined in preimmune sera, suggesting immunogenicity may be linked to the
specic glycan structure and size. Recently, Hyp-O-glycosylation of
the maize-expressed human IgA1 has been reported in which up to six
Hyp-O-arabinose residues were detected in the hinge region of this
recombinant IgG (Karnoup et al., 2005). So far, it is not yet known if
such modication has any impact on the stability, biological activity,

293

or efcacy of the IgA1 molecule, or whether these sugars could be a


source of immunogenicity or allergenicity in patients. Reports of
the presence of Hyp-O-glycosylation on plant-produced therapeutics
are likely to increase as plant-based protein production technologies
advance. Since O-glycosylation occurs mainly in the Golgi apparatus,
targeting the expression of foreign proteins to the ER would avoid this
modication. However, O-glycosylation does not necessarily convey a
negative impact on plant cell-expressed proteins. Further research is
needed to fully understand the impact of O-glycosylation on yields
and protein stability in planta as well as the half-life and pharmacodynamics of therapeutic proteins in the host (Gomord et al., 2010).
4.2. Genetic instability
The tendency towards genetic instability of suspended plant cells
poses another challenge to plant cell culture technology. Although
dedifferentiated plant cells lack of fully functional plasmodesmata
has reduced systemic post-transcriptional gene silencing (PTGS) and
are morphologically stable (Offringa et al., 1990; Shih and Doran,
2009), they have been frequently shown to suffer from genetic
instability. The underlying causes of this instability may result from
somaclonal variation (gene drift) (Cote et al., 2001), transgene loss
(Kononowicz et al., 1990; Ulker et al., 1999), transcriptional gene
silencing (Bruening, 1998; Chandler and Vaucheret, 2001; Weld et al.,
2001), or other undesirable genetic changes. Epigenetic transcriptional silencing is considered the dominant contributing factor to the
unstable expression of transgenes in plant cells that are maintained in
culture for extended periods (Matzke and Matzke, 1998; Phillips et al.,
1994). Transcriptional silencing is associated with meiotically
heritable epigenetic modications often, but not always, through
cytosine methylation (Matzke and Matzke, 1998). Due to genetic
instability, plant cell cultures will lose the capacity for high production
levels as cells with suppressed protein expression capacity begin to
dominate the culture (Gao et al., 1991; James and Lee, 2006; Shih and
Doran, 2009). For example, the production of a mAb (IgG1) has been
found to decline severely in tobacco cell cultures maintained for a
period of 3 years (Sharp and Doran, 2001b) in comparison with
relatively constant levels of this antibody expressed in tobacco hairy
root cultures over this same time period. In another example, hGMCSF protein production in tobacco NT-1 cell culture was shown to
decrease by more than 80% following 250 subculture events (James
and Lee, 2006) despite the NT-1 cell line itself being relatively stable.
Development of efcient techniques to preserve the elite cell lines,
namely cell banking, is considered a surmountable means of
overcoming this issue of genetic instability in plant cells. In general,
cell banking is considered a prerequisite for the reliable supply of
well-dened starting material for commercial recombinant protein
production on other production platforms. A three-tiered setup
consisting of a research cell bank, a master cell bank and a working
cell bank is generally accepted as the most practical approach for the
establishment and validation of the producer cell line (Parekh et al.,
2008). Cell banking by cryopreservation of elite plant cell lines has
been studied to allow the maintenance of a stable production system (Cho et al., 2007; Schmale et al., 2006). An alternative approach
is to rescreen the high-producing plant cell lines when the reduction
of protein yield is observed, provided that a reasonable fraction of
high-producing cells is still present in the cell culture. Recently, a
mathematical model for predicting the loss of productivity over
successive generations in plant cell culture has been developed
(James and Lee, 2006). This model presents a useful tool for
characterizing the stability of production cell lines and in planning
storage and rescreening efforts to ensure propagation of the high
expressing cells. In addition, co-expression of gene silencing suppressors (Baulcombe et al., 1997; Huang and McDonald, 2009) may
also be a useful strategy in maintaining high productivity of elite plant
cell culture lines.

294

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

5. Perspective and conclusion


The utility of plants as a commercially viable production platform
for pharmaceutical proteins has been a long-standing controversy in the
biotechnology industry. In the interest of addressing increased concerns
about regulatory compliance and product safety, there has been
renewed interest in plant cell cultures as a promising production
platform (Huang and McDonald, 2009; Shih and Doran, 2009). Since
plant cell cultures are inherently simpler, cheaper and safer than mammalian cell approaches and support eukaryotic processing (e.g. complex
glycosylation) absent in microbial systems, this technology has
continued to attract attention as an alternative host system. Although
the cost for plant cell culture in bioreactors is higher than an agriculture approach, the concept of being able to secret the protein of
interest into simple culture medium offers an important opportunity
for substantial cost savings (Doran, 2006a). In fact with completion of
Phase III clinical trials of plant cell-produced human glucocerebrosidase (UPLYSO) by Protalix, many in the industry are eagerly
anticipating the commercial debut of this technology early in 2011. As
plant-produced human glucocerebrosidase becomes a successful
reality, a new era in the biopharmaceutical industry will be dened
that promises to provide growth opportunity for the plant cell culture
platform.
The plant cell culture technology has made great strides in the last
two decades due to the advances in plant molecular biology and
bioreactor process engineering. As high as 247 mg/L secreted protein
yield has been achieved with rice cell culture (McDonald et al., 2005);
large-scale plant cell culture up to a volume of 100,000 L has been
successfully performed (Fischer et al., 1999a). In addition, new
bioreactor designs and advanced culture strategies (e.g. fed-batch,
perfusion culture, and continuous/semi-continuous culture) continue
to enhance the productivity of this system and facilitate the in situ
product recovery (Huang and McDonald, 2009). In recent years
signicant progress has also been made towards utilizing low-cost,
highly efcient and safe bioreactor congurations including largescale disposable bioreactors that are capable of meeting GMP
requirements (Ducos et al., 2010). Substantial advances in downstream processing including the application of advanced chromatographic technologies could signicantly enhance overall costeffectiveness (Georgiev et al., 2009). The key areas to ensure the
advancement of this technology will be in leveraging the molecular
and process engineering approaches to further increase the foreign
protein productivity, to humanize the glycans of plant glycoproteins
and to improve post-translational protein stability. With the ultimate
goal of achieving economically-feasible yields of pharmaceutical
proteins that are structurally and functionally equivalent to their
native counterparts, systematic and concerted research efforts that
are both biologically- and engineering-based will be critical to the
success of the plant cell culture protein production platform.
Acknowledgements
This work was supported by the Plant Powered Production (P3)
Center funds provided through an NSF RII Arkansas ASSET Initiative (AR
EPSCoR) grant and the Arkansas Biosciences Institute, the major
research component of the Arkansas Tobacco Settlement Proceeds Act
of 2000.
References
Abler ML, Green PJ. Control of mRNA stability in higher plants. Plant Mol Biol 1996;32:
6378.
Allen GC, Spiker S, Thompson WF. Use of matrix attachment regions (MARs) to
minimize transgene silencing. Plant Mol Biol 2000;43:36176.
Almquist KC, McLean MD, Niu YQ, Byrne G, Olea-Popelka FC, Murrant C, et al.
Expression of an anti-botulinum toxin A neutralizing single-chain Fv recombinant
antibody in transgenic tobacco. Vaccine 2006;24:207986.

Archambault J, Volesky B, Kurz WG. Surface immobilization of plant cells. Biotechnol


Bioeng 1989;33:2939.
Archambault J, Williams RD, Perrier M, Chavarie C. Production of sanguinarine by
elicited plant cell culture III. Immobilized bioreactor cultures. J Biotechnol 1996;46:
1219.
Aspelund MT, Glatz CE. Purication of recombinant plant-made proteins from corn
extracts by ultraltration. J Membr Sci 2010;353:10310.
Aviezer D, Almon-Brill E, Shaaltiel Y, Galili G, Chertkoff R, Hashmueli S, et al. Novel
enzyme replacement therapy for Gaucher disease: ongoing Phase III clinical trial
with recombinant human glucocerebrosidase expressed in plant cells. Mol Genet
Metab 2009a;96:S134.
Aviezer D, Brill-Almon E, Shaaltiel Y, Hashmueli S, Bartfeld D, Mizrachi S, et al. A plantderived recombinant human glucocerebrosidase enzymea preclinical and phase I
investigation. PLoS ONE 2009b;4:e4792.
Bai Y, Glatz CE. Capture of a recombinant protein from unclaried canola extract using
streamline expanded bed anion exchange. Biotechnol Bioeng 2003;81:85564.
Bakker H, Bardor M, Molthoff JW, Gomord V, Elbers I, Stevens LH, et al. Galactoseextended glycans of antibodies produced by transgenic plants. Proc Natl Acad Sci U
S A 2001;98:2899904.
Bardor M, Faveeuw C, Fitchette AC, Gilbert D, Galas L, Trottein F, et al. Immunoreactivity
in mammals of two typical plant glyco-epitopes, core alpha(1, 3)-fucose and core
xylose. Glycobiology 2003;13:42734.
Basaran P, Rodriguez-Cerezo E. Plant molecular farming: opportunities and challenges.
Crit Rev Biotechnol 2008;28:15372.
Bateman KS, Congiu M, Tregear GW, Clarke AE, Anderson MA. Bacitracin signicantly
reduces degradation of peptides in plant cell cultures. Biotechnol Bioeng 1997;53:
22631.
Baulcombe, D.C., Voinnet, O., Hamilton, A.J., 1997. Enhanced transgene expression by coexpression with a suppressor of post-transcriptional gene silencing (PTGS). United
States Patent, No. 7217854.
Becerra-Arteaga A, Shuler ML. Inuence of culture medium supplementation of tobacco
NT1 cell suspension cultures on the N-glycosylation of human secreted alkaline
phosphatase. Biotechnol Bioeng 2007;97:158593.
Becerra-Arteaga A, Mason HS, Shuler ML. Production, secretion, and stability of human
secreted alkaline phosphatase in tobacco NT1 cell suspension cultures. Biotechnol
Prog 2006;22:16439.
Bisaria V, Panda A. Large-scale plant cell culture: methods, applications and products.
Curr Opin Biotechnol 1991;2:3704.
Bodeutsch T, James EA, Lee JM. The effect of immobilization on recombinant protein
production in plant cell culture. Plant Cell Rep 2001;20:5626.
Boehm R. Bioproduction of therapeutic proteins in the 21st century and the role of
plants and plant cells as production platforms. Ann NY Acad Sci 2007:12134.
Boetti H, Chevalier L, Denmat LA, Thomas D, Thomasset B. Efciency of physical (Light)
or chemical (ABA, tetracycline, CuSO4 or 2-CBSU)-stimulus-dependent gus gene
expression in tobacco cell suspensions. Biotechnol Bioeng 1999;64:1-13.
Brodelius P, Mosbach K. Immobilized plant cells. Adv Appl Microbiol 1982;28:1-26.
Bruening G. Plant gene silencing regularized. Proc Natl Acad Sci U S A 1998;95:1334951.
Cabral JMS. Cell partitioning in aqueous two-phase polymer systems. Adv Biochem Eng
Biotechnol 2007;106:15171.
Callis J. Regulation of protein degradation. Plant Cell 1995;7:84557.
Camacho FG, Gomez AC, Sobczuk TM, Grima EM. Effects of mechanical and
hydrodynamic stress in agitated, sparged cultures of Porphyridium cruentum. Proc
Biochem 2000;35:104550.
Campostrini R, Carturan G, Caniato R, Piovan A, Filippini R, Innocenti G, et al.
Immobilization of plant cells in hybrid solgel materials. J Sol-Gel Sci Technol
1996;7:8797.
Cannon JG. Inammatory cytokines in nonpathological states. News Physiol Sci
2000;15:298303.
Cappelletti, E.M., Carturan, G., Piovan, A., 1999. Production of secondary metabolites with
plant cells immobilized in a porous inorganic support. United State Patent, No.
5998162.
Cassab GI. Plant cell wall proteins. Annu Rev Plant Physiol Plant Mol Biol 1998;49:
281309.
Chan MT, Yu SM. The 3 untranslated region of a rice alpha-amylase gene functions as a
sugar-dependent mRNA stability determinant. Proc Natl Acad Sci U S A 1998;95:
65437.
Chandler VL, Vaucheret H. Gene activation and gene silencing. Plant Physiol 2001;125:
1458.
Charcosset C. Membrane processes in biotechnology: an overview. Biotechnol Adv
2006;24:48292.
Chichester JA, Haaheim LR, Yusibov V. Using plant cells as inuenza vaccine substrates.
Expert Rev Vaccin 2009;8:4938.
Cho JS, Hong SM, Joo SY, Yoo JS, Kim DI. Cryopreservation of transgenic rice suspension
cells producing recombinant hCTLA4Ig. Appl Microbiol Biotechnol 2007;73:
14706.
Choi JW, Cho GH, Byun SY, Kim DI. Integrated bioprocessing for plant cell cultures. Adv
Biochem Eng Biotechnol 2001;72:63-102.
Choi SM, Lee OS, Kwon SY, Kwak SS, Yu DY, Lee HS. High expression of a human
lactoferrin in transgenic tobacco cell cultures. Biotechnol Lett 2003;25:2138.
Comai L, Moran P, Maslyar D. Novel and useful properties of a chimeric plant promoter
combining CaMV 35S and MAS elements. Plant Mol Biol 1990;15:37381.
Corrado G, Karali M. Inducible gene expression systems and plant biotechnology.
Biotechnol Adv 2009;27:73343.
Cote FX, Teisson C, Perrier X. Somaclonal variation rate evolution in plant tissue culture:
contribution to understanding through a statistical approach. In Vitro Cell Dev Biol
Plant 2001;37:53942.

J. Xu et al. / Biotechnology Advances 29 (2011) 278299


Cowen, N.M., Smith, K.A., Armstrong, K., 2007. Use of regulatory sequences in transgenic
plants. United States Patent, No. 7179902.
Cox KM, Sterling JD, Regan JT, Gasdaska JR, Frantz KK, Peele CG, et al. Glycan
optimization of a human monoclonal antibody in the aquatic plant Lemna minor.
Nat Biotechnol 2006;24:15917.
Cramer CL, Weissenborn DL, Oishi KK, Grabau EA, Bennett S, Ponce E, et al. Bioproduction of
human enzymes in transgenic tobacco. Ann NY Acad Sci 1996;792:6271.
Crawford KM, Zambryski PC. Plasmodesmata signaling: many roles, sophisticated
statutes. Curr Opin Plant Biol 1999;2:3827.
Curtis, W.R., 2004. Growing cells in a reservoir formed of a exible sterile plastic liner.
United States Patent. No. 6709862.
Curtis WR, Emery AH. Plant cell suspension culture rheology. Biotechnol Bioeng
1993;42:5206.
Daniell T, Edwards R. Changes in protein methylation associated with the elicitation
response in cell-cultures of alfalfa (Medicago sativa L). FEBS Lett 1995;360:5761.
Daniell H, Singh ND, Mason H, Streateld SJ. Plant-made vaccine antigens and
biopharmaceuticals. Trends Plant Sci 2009;14:66979.
Datla RSS, Bekkaoui F, Hammerlindl JK, Pilate G, Dunstan DI, Crosby WL. Improved highlevel constitutive foreign gene-expression in plants using an Amv Rna4
untranslated leader sequence. Plant Sci 1993;94:13949.
De Dobbeleer C, Cloutier M, Fouilland M, Legros R, Jolicoeur M. A high-rate perfusion
bioreactor for plant cells. Biotechnol Bioeng 2006;95:112637.
De Jaeger G, De Wilde C, Eeckhout D, Fiers E, Depicker A. The plantibody approach:
expression of antibody genes in plants to modulate plant metabolism or to obtain
pathogen resistance. Plant Mol Biol 2000;43:41928.
De Muynck B, Navarre C, Boutry M. Production of antibodies in plants: status after
twenty years. Plant Biotechnol J 2010;8:52963.
De Rocher EJ, Vargo-Gogola TC, Diehn SH, Green PJ. Direct evidence for rapid
degradation of Bacillus thuringiensis toxin mRNA as a cause of poor expression in
plants. Plant Physiol 1998;117:144561.
De Wilde C, De Rycke R, Beeckman T, De Neve M, Van Montagu M, Engler G, et al.
Accumulation pattern of IgG antibodies and F-ab fragments in transgenic
Arabidopsis thaliana plants. Plant Cell Physiol 1998;39:63946.
Des Molles DV, Gomord V, Bastin M, Faye L, Courtois D. Expression of a carrot invertase
gene in tobacco suspension cells cultivated in batch and continuous culture
conditions. J Biosci Bioeng 1999;87:3026.
Desai P, Shrivastava N, Padh H. Production of heterologous protein in plants: strategies
for optimal expression. Biotechnol Adv 2010;28:42735.
Dohi K, Nishikiori M, Tamai A, Ishikawa M, Meshi T, Mori M. Inducible virus-mediated
expression of a foreign protein in suspension-cultured plant cells. Arch Virol
2006;151:107584.
Doran PM. Design of mixing systems for plant cell suspensions in stirred reactors.
Biotechnol Prog 1999;15:31935.
Doran PM. Foreign protein production in plant tissue cultures. Curr Opin Biotechnol
2000;11:199204.
Doran PM. Foreign protein degradation and instability in plants and plant tissue
cultures. Trends Biotechnol 2006a;24:42632.
Doran PM. Loss of secreted antibody from transgenic plant tissue cultures due to surface
adsorption. J Biotechnol 2006b;122:3954.
Downing WL, Galpin JD, Clemens S, Lauzon SM, Samuels AL, Pidkowich MS, et al.
Synthesis of enzymatically active human alpha-L-iduronidase in Arabidopsis cgl
(complex glycan-decient) seeds. Plant Biotechnol J 2006;4:16981.
Ducos JP, Terrier B, Courtois D. Disposable bioreactors for plant micropropagation and
mass plant cell culture. Adv Biochem Eng Biotechnol 2010;115:89-115.
Dunlop DS, Yang XR, Lajtha A. The effect of elevated plasma phenylalanine levels on
protein synthesis rates in adult rat brain. Biochem J 1994;302(Pt 2):60110.
Dutta A, Pedersen H, Chin CK. Two-phase culture system for plant cells. Ann NY Acad Sci
1994;745:25160.
Eibl R, Eibl D. Design and use of the wave bioreactor for plant cell culture. Plant Tissue
Cult Eng 2006;6:20327.
Eibl R, Eibl D. Design of bioreactors suitable for plant cell and tissue cultures.
Phytochem Rev 2008;7:5938.
Eibl R, Werner S, Eibl D. Disposable bioreactors for plant liquid cultures at Litre-scale.
Eng Life Sci 2009;9:15664.
Eibl R, Kaiser S, Lombriser R, Eibl D. Disposable bioreactors: the current state-of-the-art
and recommended applications in biotechnology. Appl Microbiol Biotechnol
2010;86:419.
Faye L, Boulaous A, Benchabane M, Gomord W, Michaud D. Protein modications in
the plant secretory pathway: current status and practical implications in molecular
pharming. Vaccine 2005;23:17708.
Firek S, Draper J, Owen MR, Gandecha A, Cockburn B, Whitelam GC. Secretion of a
functional single-chain Fv protein in transgenic tobacco plants and cell-suspension
cultures. Plant Mol Biol 1993;23:86170.
Fischer R, Emans N, Schuster F, Hellwig S, Drossard J. Towards molecular farming in the
future: using plant-cell-suspension cultures as bioreactors. Biotechnol Appl
Biochem 1999a;30:10912.
Fischer R, Liao YC, Drossard J. Afnity-purication of a TMV-specic recombinant fullsize antibody from a transgenic tobacco suspension culture. J Immunol Methods
1999b;226:1-10.
Fischer R, Stoger E, Schillberg S, Christou P, Twyman RM. Plant-based production of
biopharmaceuticals. Curr Opin Plant Biol 2004;7:1528.
Francisco JA, Gawlak SL, Miller M, Bathe J, Russell D, Chace D, et al. Expression and
characterization of bryodin 1 and a bryodin 1-based single-chain immunotoxin
from tobacco cell culture. Bioconjugate Chem 1997;8:70813.
Franconi R, Demurtas OC, Massa S. Plant-derived vaccines and other therapeutics
produced in contained systems. Expert Rev Vaccines 2010;9:87792.

295

Frey AD, Karg SR, Kallio PT. Expression of rat beta(1, 4)-N-acetylglucosaminyltransferase III
in Nicotiana tabacum remodels the plant-specic N-glycosylation. Plant Biotechnol J
2009;7:3348.
Frigerio L, Vitale A, Lord JM, Ceriotti A, Roberts LM. Free ricin a chain, proricin, and
native toxin have different cellular fates when expressed in tobacco protoplasts.
J Biol Chem 1998;273:141949.
Fu LH, Miao YS, Lo SW, Seto TC, Sun SSM, Xu ZF, et al. Production and characterization of
soluble human lysosomal enzyme alpha-iduronidase with high activity from
culture media of transgenic tobacco BY-2 cells. Plant Sci 2009;177:66875.
Gallie DR, Tanguay RL, Leathers V. The tobacco etch viral 5'-leader and poly(a) tail are
functionally synergistic regulators of translation. Gene 1995;165:2338.
Gamborg OL, Miller RA, Ojima K. Nutrient requirements of suspension cultures of
soybean root cells. Exp Cell Res 1968;50:1518.
Ganapathi TR, Kumar GBS, Srinivas L, Revathi CJ, Bapat VA. Analysis of the limitations of
hepatitis B surface antigen expression in soybean cell suspension cultures. Plant
Cell Rep 2007;26:157584.
Gao JW, Lee JM, An GH. The stability of foreign protein production in genetically
modied plant cells. Plant Cell Rep 1991;10:5336.
Garbarino JE, Oosumi T, Belknap WR. Isolation of a polyubiquitin promoter and its
expression in transgenic potato plants. Plant Physiol 1995;109:13718.
Garcia-Casado G, Sanchez-Monge R, Chrispeels MJ, Armentia A, Salcedo G, Gomez L.
Role of complex asparagine-linked glycans in the allergenicity of plant glycoproteins. Glycobiology 1996;6:4717.
Georgiev MI, Weber J, Maciuk A. Bioprocessing of plant cell cultures for mass
production of targeted compounds. Appl Microbiol Biotechnol 2009;83:80923.
Geyer BC, Fletcher SP, Grifn TA, Lopker MJ, Soreq H, Mor TS. Translational control of
recombinant human acetylcholinesterase accumulation in plants. BMC Biotechnol
2007;7:27.
Gillet F, Roisin C, Fliniaux MA, Jacquin-Dubreuil A, Barbotin JN, Nava-Saucedo JE.
Immobilization of Nicotiana tabacum plant cell suspensions within calcium alginate
gel beads for the production of enhanced amounts of scopolin. Enzyme Microbiol
Technol 2000;26:22934.
Girard LS, Fabis MJ, Bastin M, Courtois D, Petiard V, Koprowski H. Expression of a human
anti-rabies virus monoclonal antibody in tobacco cell culture. Biochem Biophys Res
Commun 2006;345:6027.
Gomez L, Chrispeels MJ. Complementation of an Arabidopsis thaliana mutant that lacks
complex asparagine-linked glycans with the human cdna-encoding N-acetylglucosaminyltransferase-I. Proc Natl Acad Sci U S A 1994;91:182933.
Gomord V, Faye L. Posttranslational modication of therapeutic proteins in plants. Curr
Opin Plant Biol 2004;7:17181.
Gomord W, Chamberlain P, Jefferis R, Faye L. Biopharmaceutical production in plants:
problems, solutions and opportunities. Trends Biotechnol 2005;23:55965.
Gomord V, Fitchette AC, Menu-Bouaouiche L, Saint-Jore-Dupas C, Plasson C, Michaud D,
et al. Plant-specic glycosylation patterns in the context of therapeutic protein
production. Plant Biotechnol J 2010;8:56487.
Guan ZJ, Guo B, Huo YL, Guan ZP, Wei YH. Overview of expression of hepatitis B surface
antigen in transgenic plants. Vaccine 2010;28:735162.
Gustafsson C, Govindarajan S, Minshull J. Codon bias and heterologous protein
expression. Trends Biotechnol 2004;22:34653.
Gutierrez-Ortega A, Avila-Moreno F, Saucedo-Arias LJ, Sanchez-Torres C, Gomez-Lim
MA. Expression of a single-chain human interleukin-12 gene in transgenic tobacco
plants and functional studies. Biotechnol Bioeng 2004;85:73440.
Gutierrez-Ortega A, Sandoval-Montes C, Olivera-Flores TD, Santos-Argumedo L,
Gomez-Lim MA. Expression of functional interleukin-12 from mouse in transgenic
tomato plants. Transgenic Res 2005;14:87785.
Hacker DL, De Jesus M, Wurm FM. 25 years of recombinant proteins from reactorgrown cellswhere do we go from here? Biotechnol Adv 2009;27:10237.
Halweg C, Thompson WF, Spiker S. The rb7 matrix attachment region increases the
likelihood and magnitude of transgene expression in tobacco cells: a ow
cytometric study. Plant Cell 2005;17:41829.
He Z, Du X, Yao W, Dai J. Pharmaceutical proteins produced in plant bioreactor in recent
years. Afr J Biotechnol 2008;7:491725.
Hellwig S, Drossard J, Twyman RM, Fischer R. Plant cell cultures for the production of
recombinant proteins. Nat Biotechnol 2004;22:141522.
Hershberg R, Petrov DA. Selection on codon bias. Annu Rev Genet 2008;42:28799.
Hiatt A, Cafferkey R, Bowdish K. Production of antibodies in transgenic plants. Nature
1989;342:768.
Hogue RS, Lee JM, An G. Production of a foreign protein product with genetically
modied plant cells. Enzyme Microb Technol 1990;12:5338.
Holland T, Sack M, Rademacher T, Schmale K, Altmann F, Stadlmann J, et al. Optimal
nitrogen supply as a key to increased and sustained production of a monoclonal
full-size antibody in BY-2 suspension culture. Biotechnol Bioeng 2010;107:27889.
Hood EE, Witcher DR, Maddock S, Meyer T, Baszczynski C, Bailey M, et al. Commercial
production of avidin from transgenic maize: characterization of transformant,
production, processing, extraction and purication. Mol Breed 1997;3:291306.
Hooker BS, Lee JM. Cultivation of plant cells in aqueous two-phase polymer systems.
Plant Cell Rep 1990;8:546.
Hooker BS, Lee JM, An G. Cultivation of plant cells in a stirred vessel: effect of impeller
design. Biotechnol Bioeng 1990;35:296304.
Hsiao TY, Bacani FT, Carvalho EB, Curtis WR. Development of a low capital investment reactor
system: application for plant cell suspension culture. Biotechnol Prog 1999;15:11422.
Huang TK, McDonald KA. Bioreactor engineering for recombinant protein production in
plant cell suspension cultures. Biochem Bioeng J 2009;45:16884.
Huang J, Sutliff TD, Wu L, Nandi S, Benge K, Terashima M, et al. Expression and
purication of functional human alpha-1-Antitrypsin from cultured plant cells.
Biotechnol Prog 2001;17:12633.

296

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

Huang JM, Nandi S, Wu LY, Yalda D, Bartley G, Rodriguez R, et al. Expression of natural
antimicrobial human lysozyme in rice grains. Mol Breed 2002a;10:8394.
Huang JM, Wu LY, Yalda D, Adkins Y, Kelleher SL, Crane M, et al. Expression of functional
recombinant human lysozyme in transgenic rice cell culture. Transgenic Res
2002b;11:22939.
Huang LF, Liu YK, Lu CA, Hsieh SL, Yu SM. Production of human serum albumin by
sugar starvation induced promoter and rice cell culture. Transgenic Res 2005;14:
56981.
Huang TK, Plesha MA, Falk BW, Dandekar AM, McDonald KA. Bioreactor strategies for
improving production yield and functionality of a recombinant human protein in
transgenic tobacco cell cultures. Biotechnol Bioeng 2009;102:50820.
Huang TK, Plesha MA, McDonald KA. Semicontinuous bioreactor production of a
recombinant human therapeutic protein using a chemically inducible viral
amplicon expression system in transgenic plant cell suspension cultures.
Biotechnol Bioeng 2010;106:40821.
Hussack G, Grohs BM, Almquist KC, McLean MD, Ghosh R, Hall JC. Purication of plantderived antibodies through direct immobilization of afnity ligands on cellulose.
J Agric Food Chem 2010;58:34519.
Ilieva M, Pavlov A, Bacalova A. Phosphodiesterase production in an aqueous twophase system by Nicotiana tabacum 1507. Appl Biochem Biotechnol 2001;90:
26172.
Jabeen R, Khan MS, Zafar Y, Anjum T. Codon optimization of cry1Ab gene for hyper
expression in plant organelles. Mol Biol Rep 2010;37:10117.
James E, Lee JM. The production of foreign proteins from genetically modied plant
cells. Advances in biochemical engineering, biotechnology. In: Zhong JJ, editor.
Advances in Biochemical Engineering /BiotechnologyPlant Cells, vol. 72. Berlin:
Springer; 2001. p. 12756.
James E, Lee JM. Loss and recovery of protein productivity in genetically modied plant
cell lines. Plant Cell Rep 2006;25:7237.
James EA, Wang CL, Wang ZP, Reeves R, Shin JH, Magnuson NS, et al. Production and
characterization of biologically active human GM-CSF secreted by genetically
modied plant cells. Protein Expr Purif 2000;19:1318.
James E, Mills DR, Lee JM. Increased production and recovery of secreted foreign
proteins from plant cell cultures using an afnity chromatography bioreactor.
Biochem Bioeng J 2002;12:20513.
Jo SH, Kwon SY, Park DS, Yang KS, Kim JW, Lee KT, et al. High-yield production of
functional human lactoferrin in transgenic cell cultures of Siberian ginseng
(Acanthopanax senticosus). Biotechnol Bioprocess Eng 2006;11:4428.
Johnson M, Lanthier S, Massie B, Lefebvre G, Kamen AA. Use of the Centritech lab
centrifuge for perfusion culture of hybridoma cells in protein-free medium.
Biotechnol Prog 1996;12:85564.
Joo SY, Lee KH, Lee YI, Kim DI. Enhanced production of hGM-CSF by medium exchange
in transgenic Oryza sativa L. suspension cultures. Enzyme Microbiol Technol
2006;39:4869.
Jose W, Pedersen H, Chin CK. Immobilization of plant cells in a hollow-ber reactor. Ann
NY Acad Sci 1983;413:40912.
Judge NA, Mason HS, O'Brien AD. Plant cell-based intimin vaccine given orally to mice
primed with intimin reduces time of Escherichia coli O157: H7 shedding in feces.
Infect Immun 2004;72:16875.
Kang TJ, Loc NH, Jang MO, Yang MS. Modication of the cholera toxin B subunit coding
sequence to enhance expression in plants. Mol Breed 2004;13:14353.
Karg SR, Kallio PT. The production of biopharmaceuticals in plant systems. Biotechnol
Adv 2009;27:87994.
Karnoup AS, Turkelson V, Anderson WHK. O-Linked glycosylation in maize-expressed
human IgA1. Glycobiology 2005;15:96581.
Kaufman RJ. Post-translational modications required for coagulation factor secretion
and function. Thromb Haemost 1998;79:106879.
Kawabe A, Miyashita NT. Patterns of codon usage bias in three dicot and four monocot
plant species. Genes Genet Syst 2003;78:34352.
Kawahara H, Mitsuda S, Kumazawa E, Takeshita Y. High-density culture of FM-3A cells
using a bioreactor with an external tangential-ow ltration device. Cytotechnology 1994;14:616.
Kawana Y, Sasamoto H. Stimulation effects of salts on growth in suspension culture of a
mangrove plant, Sonneratia alba, compared with another mangrove, Bruguiera
sexangula and non-mangrove tobacco BY-2 cells. Plant Biotechnol 2008;25:1515.
Kay R, Chan A, Daly M, McPherson J. Duplication of CaMV 35S promoter sequences
creates a strong enhancer for plant genes. Science 1987;236:1299302.
Kieliszewski MJ. The latest hype on Hyp-O-glycosylation codes. Phytochemistry
2001;57:31923.
Kieliszewski MJ, Lamport DTA. Extensin-repetitive motifs, functional sites, posttranslational codes, and phylogeny. Plant J 1994;5:15772.
Kieliszewski MJ, Shpak E. Synthetic genes for the elucidation of glycosylation codes for
arabinogalactan-proteins and other hydroxyproline-rich glycoproteins. Cell Mol
Life Sci 2001;58:138698.
Kieliszewski, M.J., Xu, J., 2006. Methods of producing peptides/proteins and peptides/
proteins produced thereby. United State Patent, Publication No. US-20060026719.
Kieliszewski, M.J., Xu, J., Kopchick, J.J., Okada, S., 2006. Glycoproteins produced in plants
and methods of their use. United State Patent. Publication No. US-20060148680.
Kieran PM, MacLoughlin PF, Malone DM. Plant cell suspension cultures: some
engineering considerations. J Biotechnol 1997;59:3952.
Kim DJ, Chang HN, Liu JR. Plant cell immobilization in a dual hollow ber bioreactor.
Biotechnol Tech 1989;3:13944.
Kim DI, Cho GH, Pedersen H, Chin CK. A hybrid bioreactor for high density cultivation of
plant cell suspensions. Appl Microbiol Biotechnol 1991;34:7269.
Kim Y, Wyslouzil BE, Weathers PJ. A comparative study of mist and bubble column
reactors in the in vitro production of artemisinin. Plant Cell Rep 2001;20:4515.

Kim TG, Kim HM, Lee HJ, Shin YJ, Kwon TH, Lee NJ, et al. Reduced protease activity in
transformed rice cell suspension cultures expressing a proteinase inhibitor. Protein
Expr Purif 2007;53:2704.
Kim TG, Baek MY, Lee EK, Kwon TH, Yang MS. Expression of human growth hormone in
transgenic rice cell suspension culture. Plant Cell Rep 2008a;27:88591.
Kim TG, Lee HJ, Jang YS, Shin YJ, Kwon TH, Yang MS. Co-expression of proteinase
inhibitor enhances recombinant human granulocyte-macrophage colony stimulating factor production in transgenic rice cell suspension culture. Protein Expr
Purif 2008b;61:11721.
Ko K, Brodzik R, Steplewski Z. Production of antibodies in plants: approaches and
perspectives. In: Karasev AV, editor. Plant-produced microbial vaccines. Berlin
Herdelberg: Springer; 2009. p. 5578.
Komarnytsky S, Borisjuk N, Yakoby N, Garvey A, Raskin I. Cosecretion of protease
inhibitor stabilizes antibodies produced by plant roots. Plant Physiol 2006;141:
118593.
Kononowicz AK, Floryanowicz-Czekalska K, Clithero J, Meyers A, Hasegawa PM, Bressan
RA. Chromosome number and DNA content of tobacco cells adapted to NaCl. Plant
Cell Rep 1990;8:6725.
Koprivova A, Stemmer C, Altmann F, Hoffmann A, Kopriva S, Gorr G, et al. Targeted
knockouts of Physcomitrella lacking plantspecic immunogenic N-glycans. Plant
Biotechnol J 2004;2:51723.
Koyama K, Seki M. Cultivation of yeast and plant cells entrapped in the low-viscous
liquid-core of an alginate membrane capsule prepared using polyethylene glycol.
J Biosci Bioeng 2004;97:1118.
Kumar GBS, Ganapathi TR, Revathi CJ, Srinivas L, Bapat VA. Expression of hepatitis B
surface antigen in transgenic banana plants. Planta 2005;222:48493.
Kumar GBS, Ganapathi TR, Srinivas L, Revathi CJ, Bapat VA. Hepatitis B surface antigen
expression in NT-1 cells of tobacco using different expression cassettes. Biol Plant
2007;51:46771.
Kwon TH, Kim YS, Lee JH, Yang MS. Production and secretion of biologically active
human granulocyte-macrophage colony stimulating factor in transgenic tomato
suspension cultures. Biotechnol Lett 2003a;25:15714.
Kwon TH, Seo JE, Kim J, Lee JH, Jang YS, Yang MS. Expression and secretion of the
heterodimeric protein interleukin-12 in plant cell suspension culture. Biotechnol
Bioeng 2003b;81:8705.
Kwon TH, Shin YM, Kim YS, Jang YS, Yang MS. Secretory production of hGM-CSF with a
high specic biological activity by transgenic plant cell suspension culture.
Biotechnol Bioprocess Eng 2003c;8:13541.
LaCount W, An GH, Lee JM. The effect of polyvinylpyrrolidone (PVP) on the heavy chain
monoclonal antibody production from plant suspension cultures. Biotechnol Lett
1997;19:936.
Lai HF, Engle M, Fuchs A, Keller T, Johnson S, Gorlatov S, et al. Monoclonal antibody
produced in plants efciently treats West Nile virus infection in mice. Proc Natl
Acad Sci U S A 2010;107:241924.
Lam, E., Benfey, P.N., Gilmartin, P.M., Chua, N.H., 1991. Promoter enhancer element for
gene expression in plant roots. United States Patent, No. 5023179.
Larrick JW, Yu L, Naftzger C, Jaiswal S, Wycoff K. Production of secretory IgA antibodies
in plants. Biomol Eng 2001;18:8794.
Lau OS, Sun SSM. Plant seeds as bioreactors for recombinant protein production.
Biotechnol Adv 2009;27:101522.
Leader B, Baca QJ, Golan DE. Protein therapeutics: a summary and pharmacological
classication. Nat Rev Drug Discov 2008;7:2139.
Lee SY, Kim DI. Stimulation of murine granulocyte macrophage-colony stimulating
factor production by Pluronic F-68 and polyethylene glycol in transgenic Nicotiana
tabacum cell culture. Biotechnol Lett 2002;24:177983.
Lee SY, Kim DI. Perfusion cultivation of transgenic Nicotiana tabacum suspensions in
bioreactor for recombinant protein production. J Microbiol Biotechnol 2006;16:
6737.
Lee, J.M., Magnuson, N.S., An, G., Reeves, R., 2000. Production of secreted foreign
polypeptides in plant cell culture. United States Patent, No. 6020169.
Lee JH, Kim NS, Kwon TH, Jang YS, Yang MS. Increased production of human
granulocyte-macrophage colony stimulating factor (hGM-CSF) by the addition of
stabilizing polymer in plant suspension cultures. J Biotechnol 2002;96:20511.
Lee JH, Loc NH, Kwon TH, Yang MS. Partitioning of recombinant human granulocytemacrophage colony stimulating factor (hGM-CSF) from plant cell suspension
culture in PEG/sodium phosphate aqueous two-phase systems. Biotechnol
Bioprocess Eng 2004a;9:126.
Lee SY, Kim YH, Roh YS, Myoung HJ, Lee KY, Kim DI. Bioreactor operation for transgenic
Nicotiana tabacum cell cultures and continuous production of recombinant human
granulocyte-macrophage colony-stimulating factor by perfusion culture. Enzyme
Microb Technol 2004b;35:66371.
Lee, H.S., Kwak, S.S., Kwon, S.Y., Yu, D.Y., Kim, J.W., Lee, O.S., 2006. Method for producing
human lactoferrin in plant cell culture. United States Patent, No. 7118914.
Leonard R, Petersen BO, Himly M, Kaar W, Wopfner N, Kolarich D, et al. Two novel types
of O-glycans on the mugwort pollen allergen Art v 1 and their role in antibody
binding. J Biol Chem 2005;280:793240.
Lerouge P, Cabanes-Macheteau M, Rayon C, Fischette-Laine AC, Gomord V, Faye L. Nglycoprotein biosynthesis in plants: recent developments and future trends. Plant
Mol Biol 1998;38:3148.
Li HJ, d'Anjou M. Pharmacological signicance of glycosylation in therapeutic proteins.
Curr Opin Biotechnol 2009;20:67884.
Lienard D, Dinh OT, van Oort E, Van Overtvelt L, Bonneau C, Wambre E, et al.
Suspension-cultured BY-2 tobacco cells produce and mature immunologically
active house dust mite allergens. Plant Biotechnol J 2007;5:93-108.
Linsmayer EM, Skoog F. Organic growth factor require- ments of tobacco tissue cultures.
Physiol Plant 1965;18:10027.

J. Xu et al. / Biotechnology Advances 29 (2011) 278299


Liu QP, Xue QZ. Comparative studies on codon usage pattern of chloroplasts and their
host nuclear genes in four plant species. J Genet 2005;84:5562.
Liu YK, Seki M, Furusaki S. Plant cell immobilization in loofa sponge using two-way
bubble circular system. J Chem Eng Jpn 1999;32:8-14.
Liu S, Bugos RC, Dharmasiri N, Su WW. Green uorescent protein as a secretory reporter
and a tool for process optimization in transgenic plant cell cultures. J Biotechnol
2001;87:1-16.
Ma JKC, Drake PMW, Christou P. The production of recombinant pharmaceutical
proteins in plants. Nat Rev Genet 2003;4:794805.
Ma SW, Huang Y, Davis A, Yin ZQ, Mi QS, Menassa R, et al. Production of biologically
active human interleukin-4 in transgenic tobacco and potato. Plant Biotechnol J
2005;3:30918.
Magnuson NS, Linzmaier PM, Gao JW, Reeves R, An GH, Lee JM. Enhanced recovery of a
secreted mammalian protein from suspension culture of genetically modied
tobacco cells. Protein Expr Purif 1996;7:2208.
Magnuson NS, Linzmaier PM, Reeves R, An GH, HayGlass K, Lee JM. Secretion of
biologically active human interleukin-2 and interleukin-4 from genetically
modied tobacco cells in suspension culture. Protein Expr Purif 1998;13:4552.
Matsumoto S, Ishii A, Ikura K, Ueda M, Sasaki R. Expression of human erythropoietin in
cultured tobacco cells. Biosci Biotechnol Biochem 1993;57:124952.
Matsuo K, Hong JS, Tabayashi N, Ito A, Masuta C, Matsumura T. Development of
Cucumber mosaic virus as a vector modiable for different host species to produce
therapeutic proteins. Planta 2007;225:27786.
Matzke AJM, Matzke MA. Position effects and epigenetic silencing of plant transgenes.
Curr Opin Plant Biol 1998;1:1428.
McDonald KA, Hong LM, Trombly DM, Xie Q, Jackman AP. Production of human alpha-1antitrypsin from transgenic rice cell culture in a membrane bioreactor. Biotechnol
Prog 2005;21:72834.
Medrano G, Reidy MJ, Liu J, Ayala J, Dolan MC, Cramer CL. Rapid system for evaluating
bioproduction capacity of complex pharmaceutical proteins in plants. Methods Mol
Biol 2009;483:5167.
Medrano G, Stephens N, McMickle A, Dolan M, Radin D, Erf G, et al. Efcient plant-based
production of chicken IL-12 yields a strong immunostimulatory cytokine. J
Interferon Cytokine Res 2010;30:2131.
Menkhaus TJ, Bai Y, Zhang CM, Nikolov ZL, Glatz CE. Considerations for the recovery of
recombinant proteins from plants. Biotechnol Prog 2004;20:100114.
Mihaliak CA, Webb SR. Plant-cell-produced vaccines for animal health. Feedinfo News
Serv 2005;9:14.
Miller RA, Shyluk JP, Gamborg OL, Kirkpatrick JW. Phytostat for continuous culture and
automatic sampling of plant-cell suspensions. Science 1968;159:5402.
Min JY, Jung HY, Kang SM, Kim YD, Kang YM, Park DJ, et al. Production of tropane
alkaloids by small-scale bubble column bioreactor cultures of Scopolia parviora
adventitious roots. Bioresour Technol 2007;98:174853.
Mishra S, Yadav DK, Tuli R. Ubiquitin fusion enhances cholera toxin B subunit
expression in transgenic plants and the plant-expressed protein binds GM1
receptors more efciently. J Biotechnol 2006;127:95-108.
Murashige T, Skoog F. A revised medium for rapid growth and bioassays with tobacco
cultures. Physiol Plant 1962;15:47397.
Murray EE, Lotzer J, Eberle M. Codon usage in plant genes. Nucleic Acids Res 1989;17:47798.
Namdev PK, Dunlop EH. Shear sensitivity of plant-cells in suspensionspresent and
future. Appl Biochem Biotechnol 1995;54:10931.
Nikolov ZL, Woodard SL. Downstream processing of recombinant proteins from
transgenic feedstock. Curr Opin Biotechnol 2004;15:47986.
Nuttall J, Vine N, Hadlington JL, Drake P, Frigerio L, Ma JKC. ER-resident chaperone
interactions with recombinant antibodies in transgenic plants. Eur J Biochem
2002;269:604251.
Offringa R, de Groot MJ, Haagsman HJ, Does MP, van den Elzen PJ, Hooykaas PJ.
Extrachromosomal homologous recombination and gene targeting in plant cells
after Agrobacterium mediated transformation. EMBO J 1990;9:307784.
Omar R, Abdullah MA, Hasan MA, Marziah M. Development of growth medium for
Centella asiatica cell culture via response surface methodology. Am J Appl Sci
2004;1:2159.
Osuna L, Moyano E, Mangas S, Bonll M, Cusido RM, Pinol MT, et al. Immobilization of
Galphimia glauca plant cell suspensions for the production of enhanced amounts of
galphimine-B. Planta Med 2008;74:949.
Ozawa K, Takaiwa F. Highly efcient Agrobacterium-mediated transformation of
suspension-cultured cell clusters of rice (Oryza sativa L.). Plant Sci 2010;179:3337.
Padidam M. Chemically regulated gene expression in plants. Curr Opin Plant Biol
2003;6:16977.
Panahi M, Cheng XY, Alli Z, Sardana R, Callaghan M, Phipps J, et al. Plant-derived
recombinant human insulin-like growth factor precursor prohormone IGF-1B
caused differentiation of human neuroblastoma cell lines SH-SY5Y. Mol Breed
2003;12:2131.
Panahi M, Alli Z, Cheng XY, Belbaraka L, Belgoudi J, Sardana R, et al. Recombinant
protein expression plasmids optimized for industrial E. coli fermentation and plant
systems produce biologically active human insulin-like growth factor-1 in
transgenic rice and tobacco plants. Transgenic Res 2004;13:24559.
Parekh S, Srinivasan V, Horn M. Bioprocessing using novel cell culture systems. Adv
Appl Microbiol 2008;63:10543.
Park Y, Cheong H. Expression and production of recombinant human interleukin-2 in
potato plants. Protein Expr Purif 2002;25:1605.
Park CI, Lee SJ, Kang SH, Jung HS, Kim DI, Lim SM. Fed-batch cultivation of transgenic
rice cells for the production of hCTLA4Ig using concentrated amino acids. Proc
Biochem 2010;45:6774.
Parsons J, Wirth S, Dominguez M, Bravo-Almonacid F, Giulietti AM, Rodriguez Talou J.
Production of human epidermal growth factor (hEGF) by in vitro cultures of

297

Nicotiana tabacum: effect of tissue differentiation and sodium nitroprusside


addition. Int J Biotechnol Biochem 2010;6:1318.
Peckham GD, Bugos RC, Wei WS. Purication of GFP fusion proteins from transgenic
plant cell cultures. Protein Expr Purif 2006;49:1839.
Petersen K, Leah R, Knudsen S, Cameron-Mills V. Matrix attachment regions (MARs)
enhance transformation frequencies and reduce variance of transgene expression
in barley. Plant Mol Biol 2002;49:4558.
Petruccelli S, Otegui MS, Lareu F, Dinh OT, Fitchette AC, Circosta A, et al. A KDEL-tagged
monoclonal antibody is efciently retained in the endoplasmic reticulum in leaves,
but is both partially secreted and sorted to protein storage vacuoles in seeds. Plant
Biotechnol J 2006;4:51127.
Phillips RL, Kaeppler SM, Olhoft P. Genetic instability of plant-tissue cultures
breakdown of normal controls. Proc Natl Acad Sci U S A 1994;91:52226.
Ponstein AS, Verwoerd TC, Pen J. Production of enzymes for industrial use. Ann NY Acad
Sci 1996;792:918.
Prakash G, Srivastava AK. Statistical media optimization for cell growth and
azadirachtin production in Azadirachta indica (A. Juss) suspension cultures. Proc
Biochem 2005;40:3795800.
Ramirez N, Lorenzo D, Palenzuela D, Herrera L, Ayala M, Fuentes A, et al. Single-chain
antibody fragments specic to the hepatitis B surface antigen, produced in
recombinant tobacco cell cultures. Biotechnol Lett 2000;22:12336.
Ramirez N, Oramas P, Ayala M, Rodriguez M, Perez M, Gavilondo J. Expression and longterm stability of a recombinant single-chain Fv antibody fragment in transgenic
Nicotiana tabacum seeds. Biotechnol Lett 2001;23:479.
Ramirez N, Ayala M, Lorenzo D, Palenzuela D, Herrera L, Doreste V, et al. Expression of a
single-chain Fv antibody fragment specic for the Hepatitis B surface antigen in
transgenic tobacco plants. Transgenic Res 2002;11:614.
Ramirez N, Rodriguez M, Ayala M, Cremata J, Perez M, Martinez A, et al. Expression and
characterization of an anti-(hepatitis B surface antigen) glycosylated mouse antibody
in transgenic tobacco (Nicotiana tabacum) plants and its use in the immunopurication of its target antigen. Biotechnol Appl Biochem 2003;38:22330.
Rao SR, Ravishankar GA. Plant cell cultures: chemical factories of secondary
metabolites. Biotechnol Adv 2002;20:10153.
Ritala A, Wahlstrom EH, Holkeri H, Hafren A, Makelainen K, Baez J, et al. Production of a
recombinant industrial protein using barley cell cultures. Protein Expr Purif 2008;59:
27481.
Roberts SC, Shuler ML. Large-scale plant cell culture. Curr Opin Biotechnol 1997;8:1549.
Roja G, Bhangale AS, Juvekar AR, Eapen S, D'Souza SF. Enhanced production of the
polysaccharide arabinogalactan using immobilized cultures of Tinospora cordifolia
by elicitation and in situ adsorption. Biotechnol Prog 2005;21:168891.
Ruggiero F, Exposito JY, Bournat P, Gruber V, Perret S, Comte J, et al. Triple helix
assembly and processing of human collagen produced in transgenic tobacco plants.
FEBS Lett 2000;469:1326.
Russell, D.R., Fuller, J.T. 2000. Method for producing antibodies in plant cells. United State
Patent, No. 6080560.
Rybicki EP. Plant-made vaccines for humans and animals. Plant Biotechnol J 2010;8:
62037.
Sahai OP, Shuler ML. Multistage continuous culture to examine secondary metabolite
formation in plant cells: phenolics from Nicotiana tabacum. Biotechnol Bioeng
1984;26:2736.
Sainsbury F, Lomonossoff GP. Extremely high-level and rapid transient protein production
in plants without the use of viral replication. Plant Physiol 2008;148:12128.
Sainsbury F, Lavoie PO, D'Aoust MA, Vezina LP, Lomonossoff GP. Expression of multiple
proteins using full-length and deleted versions of cowpea mosaic virus RNA-2.
Plant Biotechnol J 2008;6:8292.
Sajc L, Grubisic D, Vunjak-Novakovic G. Bioreactors for plant engineering: an outlook
for further research. Biochem Bioeng J 2000;4:8999.
Samalova M, Brzobohaty B, Moore I. pOp6/LhGR: a stringently regulated and highly
responsive dexamethasone-inducible gene expression system for tobacco. Plant J
2005;41:91935.
Schenk RU, Hildebrandt AC. Medium and techniques for induction and growth of
monocotyledonous and dicotyledonous plant cell cultures. Can J Bot 1972;50:
199204.
Schiermeyer A, Schinkel H, Apel S, Fischer R, Schillberg S. Production of Desmodus
rotundus salivary plasminogen activator alpha1 (DSPAalpha1) in tobacco is
hampered by proteolysis. Biotechnol Bioeng 2005;89:84858.
Schillberg S, Fischer R, Emans N. Molecular farming of recombinant antibodies in plants.
Cell Mol Life Sci 2003;60:43345.
Schinkel H, Schiermeyer A, Soeur R, Fischer R, Schillberg S. Production of an active
recombinant thrombomodulin derivative in transgenic tobacco plants and
suspension cells. Transgenic Res 2005;14:2519.
Schmale K, Rademacher T, Fischer R, Hellwig S. Towards industrial usefulnesscryocell-banking of transgenic BY-2 cell cultures. J Biotechnol 2006;124:30211.
Semenyuk EG, Stremovskii OA, Orlova IV, Balandin TG, Nosov AM, Buryanov YI, et al.
Biosynthesis of the scFv antibody to human ferritin in plant and bacterial
producers. Mol Biol 2003;37:7806.
Shaaltiel Y, Bartfeld D, Hashmueli S, Baum G, Brill-Almon E, Galili G, et al. Production of
glucocerebrosidase with terminal mannose glycans for enzyme replacement therapy
of Gaucher's disease using a plant cell system. Plant Biotechnol J 2007;5:57990.
Shaaltiel, Y., Hashmueli, S., Bartfeld, D., Baum, G., Ratz, T., Mizrachi, E. et al., 2009. System
and method for production of antibodies in plant cell culture. United States Patent.
Publication No. 20090082548.
Sharm MK, Jani D, Thungapathra M, Gautam JK, Meena LS, Singh Y, et al. Expression of
accessory colonization factor subunit A (ACFA) of Vibrio cholerae and ACFA fused to
cholera toxin B subunit in transgenic tomato (Solanum lycopersicum). J Biotechnol
2008;135:227.

298

J. Xu et al. / Biotechnology Advances 29 (2011) 278299

Sharma AK, Sharma MK. Plants as bioreactors: recent developments and emerging
opportunities. Biotechnol Adv 2009;27:81132.
Sharma N, Kim TG, Yang MS. Production and secretion of human interleukin-18 in
transgenic tobacco cell suspension culture. Biotechnol Bioprocess Eng 2006;11:1549.
Sharp JM, Doran PM. Characterization of monoclonal antibody fragments produced by
plant cells. Biotechnol Bioeng 2001a;73:33846.
Sharp JM, Doran PM. Strategies for enhancing monoclonal antibody accumulation in
plant cell and organ cultures. Biotechnol Prog 2001b;17:97992.
Shibasaki N, Hirose K, Yonemoto T, Takadi T. Suspension culture of Nicotiana tabacum
cells in a rotary-drum bioreactor. J Chem Technol Biotechnol 1992;53:35963.
Shih SMH, Doran PM. Foreign protein production using plant cell and organ cultures:
advantages and limitations. Biotechnol Adv 2009;27:103642.
Shin YJ, Hong SY, Kwon TH, Jang YS, Yang MS. High level of expression of recombinant
human granulocyte-macrophage colony stimulating factor in transgenic rice cell
suspension culture. Biotechnol Bioeng 2003;82:77883.
Shin YJ, Chong YJ, Han KB, Yang MS, Kwon TH. N-linked glycan analysis of glycoproteins
secreted from rice cell suspension cultures under sugar starvation. Enzyme Microb
Technol 2010a;47:18993.
Shin YJ, Lee NJ, Kim J, An XH, Yang MS, Kwon TH. High-level production of bioactive
heterodimeric protein human interleukin-12 in rice. Enzyme Microb Technol
2010b;46:34751.
Showalter AM. Structure and function of plant cell wall proteins. Plant Cell 1993;5:9-23.
Shpak E, Leykam JF, Kieliszewski MJ. Synthetic genes for glycoprotein design and the
elucidation of hydroxyproline-O-glycosylation codes. Proc Natl Acad Sci U S A 1999;96:
1473641.
Shuler ML, Hallsby GA, Pyne J, J.W., Cho T. Bioreactors for immobilized plant cell
cultures. Ann NY Acad Sci 1986;469:2708.
Sijmons PC, Dekker BM, Schrammeijer B, Verwoerd TC, van den Elzen PJ, Hoekema A.
Production of correctly processed human serum albumin in transgenic plants.
Biotechnology 1990;8:21721.
Sim SJ, Chang HN. Fed-batch hairy root cultures with in situ separation. Biotechnol
Bioprocess Eng 1999;4:10611.
Singh V. Disposable bioreactor for cell culture using wave-induced agitation.
Cytotechnology 1999;30:14958.
Smetanska I. Production of secondary metabolites using plant cell cultures. Food
Biotechnol 2008;111:187228.
Smith ML, Mason HS, Shuler ML. Hepatitis B surface antigen (HBsAg) expression in
plant cell culture: kinetics of antigen accumulation in batch culture and its
intracellular form. Biotechnol Bioeng 2002;80:81222.
Sojikul P, Buehner N, Mason HS. A plant signal peptide-hepatitis B surface antigen
fusion protein with enhanced stability and immunogenicity expressed in plant
cells. Proc Natl Acad Sci U S A 2003;100:220914.
Sorrentino A, Schillberg S, Fischer R, Rao R, Porta R, Mariniello L. Recombinant human
tissue transglutaminase produced into tobacco suspension cell cultures is active
and recognizes autoantibodies in the serum of coeliac patients. Int J Biochem Cell
Biol 2005;37:84251.
Sourrouille C, Marquet-Blouin E, D'Aoust MA, Kiefer-Meyer MC, Seveno M, PagnySalehabadi S, et al. Down-regulated expression of plant-specic glycoepitopes in
alfalfa. Plant Biotechnol J 2008;6:70221.
Sowana DD, Williams DRG, Dunlop EH, Dally BB, O'Neill BK, Fletcher DF. Turbulent
shear stress effects on plant cell suspension cultures. Chem Eng Res Des 2001;79:
86775.
Staub JM, Garcia B, Graves J, Hajdukiewicz PTJ, Hunter P, Nehra N, et al. High-yield
production of a human therapeutic protein in tobacco chloroplasts. Nat Biotechnol
2000;18:3338.
Stevens LH, Stoopen GM, Elbers IJW, Molthoff JW, Bakker HAC, Lommen A, et al. Effect
of climate conditions and plant developmental stage on the stability of antibodies
expressed in transgenic tobacco. Plant Physiol 2000;124:17382.
Stoger E, Sack M, Fischer R, Christou P. Plantibodies: applications, advantages and
bottlenecks. Curr Opin Biotechnol 2002a;13:1616.
Stoger E, Sack M, Perrin Y, Vaquero C, Torres E, Twyman RM, et al. Practical
considerations for pharmaceutical antibody production in different crop systems.
Mol. Breed. 2002b;9:14958.
Strasser R, Altmann F, Mach L, Glossl J, Steinkellner H. Generation of Arabidopsis thaliana
plants with complex N-glycans lacking beta 1, 2-linked xylose and core alpha 1, 3-linked
fucose. FEBS Lett. 2004;561:1326.
Strasser R, Stadlmann J, Schahs M, Stiegler G, Quendler H, Mach L, et al. Generation of
glyco-engineered Nicotiana benthamiana for the production of monoclonal
antibodies with a homogeneous human-like N-glycan structure. Plant Biotechnol.
J. 2008;6:392402.
Streateld SJ. Approaches to achieve high-level heterologous protein production in
plants. Plant Biotechnol. J. 2007;5:2-15.
Su WW. Bioprocessing technology for plant-cell suspension-cultures. Appl. Biochem.
Biotechnol. 1995;50:189230.
Su WW, Arias R. Continuous plant cell perfusion culture: bioreactor characterization
and secreted enzyme production. J. Biosci. Bioeng. 2003;95:1320.
Su WW, Lee KT. Plant cell and hairy-root cultures-process characteristics, products, and
application. In: Yang ST, editor. Bioprocessing for value-added products from
renewable resources. Amsterdam, The Netherlands: Elsevier Science; 2007.
p. 26392.
Su WW, He BJ, Liang H, Sun S. A perfusion air-lift bioreactor for high density plant cell
cultivation and secreted protein production. J. Biotechnol. 1996;50:22533.
Sun X, Linden JC. Shear stress effects on plant cell suspension cultures in a rotating wall
vessel bioreactor. J. Ind. Microbiol. Biotechnol. 1999;22:447.
Tan L, Leykam JF, Kieliszewski MJ. Glycosylation motifs that direct arabinogalactan
addition to arabinogalactan-proteins. Plant Physiol. 2003;132:13629.

Tanaka H, Nishijima F, Suwa M, Iwamoto T. Rotating drum fermentor for plant cell
suspension cultures. Biotechnol. Bioeng. 1983;25:235970.
Tang JF, Wang ZZ. Recent progress on transgenic plants expressing pharmaceutical
proteins. Prog. Mod. Biomed. 2006;6:736.
Tang W, Luo XY, Samuels V. Regulated gene expression with promoters responding to
inducers. Plant Sci. 2004;166:82734.
Tavladoraki P, Benvenuto E, Trinca S, De Martinis D, Cattaneo A, Galef P. Transgenic
plants expressing a functional single-chain Fv antibody are specically protected
from virus attack. Nature 1993;366:46972.
Terashima M, Murai Y, Kawamura M, Nakanishi S, Stoltz T, Chen L, et al. Production of
functional human alpha(1)-antitrypsin by plant cell culture. Appl. Microbiol.
Biotechnol. 1999;52:51623.
Terrier B, Courtois D, Henault N, Cuvier A, Bastin M, Aknin A, et al. Two new disposable
bioreactors for plant cell culture: the wave and undertow bioreactor and the slug
bubble bioreactor. Biotechnol. Bioeng. 2007;96:91423.
Tiwari S, Verma PC, Singh PK, Tuli R. Plants as bioreactors for the production of vaccine
antigens. Biotechnol. Adv. 2009;27:44967.
Torres E, Vaquero C, Nicholson L, Sack M, Stoger E, Drossard J, et al. Rice cell culture as
an alternative production system for functional diagnostic and therapeutic
antibodies. Transgenic Res. 1999;8:4419.
Tremblay R, Wang D, Jevnikar AM, Ma S. Tobacco, a highly efcient green bioreactor for
production of therapeutic proteins. Biotechnol. Adv. 2010;28:21421.
Trexler MM, McDonald KA, Jackman AP. Bioreactor production of human alpha(1)antitrypsin using metabolically regulated plant cell cultures. Biotechnol. Prog.
2002;18:5018.
Trexler MM, McDonald KA, Jackman AP. A cyclical semicontinuous process for
production of human alpha(1)-antitrypsin using metabolically induced plant cell
suspension cultures. Biotechnol. Prog. 2005;21:3218.
Tsoi BMY, Doran PM. Effect of medium properties and additives on antibody stability
and accumulation in suspended plant cell cultures. Biotechnol. Appl. Biochem.
2002;35:17180.
Ulker B, Allen GC, Thompson WF, Spiker S, Weissinger AK. A tobacco matrix attachment
region reduces the loss of transgene expression in the progeny of transgenic
tobacco plants. Plant J. 1999;18:25363.
Valdes R, Gomez L, Padilla S, Brito J, Reyes B, Alvarez T, et al. Large-scale purication of
an antibody directed against hepatitis B surface antigen from transgenic tobacco
plants. Biochem. Biophys. Res. Commun. 2003;308:94-100.
Van der Vyver C, Schneidereit J, Driscoll S, Turner J, Kunert K, Foyer CH. Oryzacystatin I
expression in transformed tobacco produces a conditional growth phenotype and
enhances chilling tolerance. Plant Biotechnol. J. 2003;1:10112.
van Gulik WM, ten Hoopen HJG, Heijnen JJ. The application of continuous culture for
plant cell suspensions. Enzyme Microb. Technol. 2001;28:796805.
Wahl MF, An GH, Lee JM. Effects of dimethyl-sulfoxide on heavy-chain monoclonalantibody production from plant-cell culture. Biotechnol. Lett. 1995;17:4638.
Walter P. Personalised medicineplant produced vaccine tackles cancer. Chem. Ind.
Lond. 2008;15: 77.
Wang L, Coppel RL. Oral vaccine delivery: can it protect against non-mucosal
pathogens? Expert Rev. Vaccin. 2008;7:72938.
Wang GR, Qi NM. Perfusion culture of Glycyrrhiza inata suspension cells in a stir-tank
bioreactor. Aust. J. Bot. 2009;57:2406.
Wang SJ, Zhong JJ. A novel centrifugal impeller bioreactor.1. Fluid circulation, mixing,
and liquid velocity proles. Biotechnol. Bioeng. 1996a;51:5119.
Wang SJ, Zhong JJ. A novel centrifugal impeller bioreactor.2. Oxygen transfer and power
consumption. Biotechnol. Bioeng. 1996b;51:5207.
Wang XG, Zhang GH, Liu CX, Zhang YH, Xiao CZ, Fang RX. Puried cholera toxin B subunit
from transgenic tobacco plants possesses authentic antigenicity. Biotechnol. Bioeng.
2001;72:4904.
Wang DJ, Brandsma M, Yin ZQ, Wang AM, Jevnikar AM, Ma SW. A novel platform for
biologically active recombinant human interleukin-13 production. Plant Biotechnol. J. 2008;6:50415.
Wang GR, Qi NM, Wang ZM. Application of a stir-tank bioreactor for perfusion culture
and continuous harvest of Glycyrrhiza inata suspension cells. Afr. J. Biotechnol.
2010;9:34751.
Weathers PJ, Hemmavanh DD, Walcerz DB, Cheetham RD, Smith TC. Interactive effects
of nitrate and phosphate salts, sucrose, and inoculum culture age on growth and
sesquiterpene production in Artemisia annua hairy root cultures. In Vitro Cell Dev.
Biol. Plant 1997;33:30612.
Weathers PJ, Towler MJ, Xu JF. Bench to batch: advances in plant cell culture for
producing useful products. Appl. Microbiol. Biotechnol. 2010;85:133951.
Weld R, Heinemann J, Eady C. Transient GFP expression in Nicotiana plumbaginifolia
suspension cells: the role of gene silencing, cell death and T-DNA loss. Plant Mol.
Biol. 2001;45:37785.
Willats WGT, Marcus SE, Knox JP. Generation of a monoclonal antibody specic to (1 -N5)alpha-L-arabinan. Carbohydr. Res. 1998;308:14952.
Wilson IBH, Harthill JE, Mullin NP, Ashford DA, Altmann F. Core alpha 1, 3-fucose is a key part
of the epitope recognized by antibodies reacting against plant N-linked oligosaccharides
and is present in a wide variety of plant extracts. Glycobiology 1998;8:65161.
Wirth S, Calamante G, Mentaberry A, Bussmann L, Lattanzi M, Baranao L, et al. Expression of
active human epidermal growth factor (hEGF) in tobacco plants by integrative and
non-integrative systems. Mol. Breed. 2004;13:2335.
Wongsamuth R, Doran PM. Production of monoclonal antibodies by tobacco hairy roots.
Biotechnol. Bioeng. 1997;54:40115.
Woodnutt G, Violand B, North M. Advances in protein therapeutics. Curr. Opin. Drug
Discov. Dev. 2008;11:75461.
Wu HZ, Scissum-Gunn K, Singh NK, Giambrone JJ. Toward the development of a plantbased vaccine against reovirus. Avian Dis. 2009;53:37681.

J. Xu et al. / Biotechnology Advances 29 (2011) 278299


Xu JF, Yin PQ, Wei XG, Su ZG. Self-immobilized aggregated culture of Taxus cuspidate for
improved taxol production. Biotechnol. Tech. 1998a;12:2414.
Xu JF, Ying PQ, Han AM, Su ZG. Enhanced salidroside production in liquid-cultivated
compact callus aggregates of Rhodiola sachalinensis: manipulation of plant growth
regulators and sucrose. Plant Cell Tissue Org. Cult. 1998b;55:538.
Xu JF, Shpak E, Gu TY, Moo-Young M, Kieliszewski M. Production of recombinant plant
gum with tobacco cell culture in bioreactor and gum characterization. Biotechnol.
Bioeng. 2005;90:57888.
Xu JF, Tan L, Goodrum KJ, Kieliszewski MJ. High-yields and extended serum half-life of
human interferon alpha 2b expressed in tobacco cells as Arabinogalactan-protein
fusions. Biotechnol. Bioeng. 2007;97:997-1008.
Xu JF, Tan L, Lamport DTA, Showalter AM, Kieliszewski MJ. The O-Hyp glycosylation
code in tobacco and Arabidopsis and a proposed role of Hyp-glycans in secretion.
Phytochem. 2008;69:163140.
Xu JF, Okada S, Tan L, Goodrum KJ, Kopchick JJ, Kieliszewski MJ. Human growth
hormone expressed in tobacco cells as an arabinogalactan-protein fusion
glycoprotein has a prolonged serum life. Transgenic Res. 2010;19(5):84967.
Xu ZF, Teng WL, Chye ML. Inhibition of endogenous trypsin- and chymotrypsin-like
activities in transgenic lettuce expressing heterogeneous proteinase inhibitor
SaPIN2a. Planta 2004;218:6239.
Yang JJ, Barr LA, Fahnestock SR, Liu ZB. High yield recombinant silk-like protein production
in transgenic plants through protein targeting. Transgenic Res. 2005;14:31324.
Yano A, Maeda F, Takekoshi M. Transgenic tobacco cells producing the human monoclonal
antibody to hepatitis B virus surface antigen. J. Med. Virol. 2004;73:20815.
Yano A, Takekoshi M, Morita E, Imai S, Nisizawa T, Hanada N. Production of fab fragment
corresponding to surface protein antigen of Streptococcus mutans serotype c-derived
peptide by Escherichia coli and cultured tobacco cells. J. Biosci. Bioeng. 2006;101:2516.

299

Yasuda H, Hayashi Y, Jomori T, Takaiwa F. The correlation between expression and localization of a foreign gene product in rice endosperm. Plant Cell Physiol. 2006;47:75663.
Yates EA, Valdor JF, Haslam SM, Morris HR, Dell A, Mackie W, et al. Characterization of
carbohydrate structural features recognized by anti-arabinogalactan-protein
monoclonal antibodies. Glycobiology 1996;6:1319.
Yu J, Langridge WHR. A plant-based multicomponent vaccine protects mice from
enteric diseases. Nat. Biotechnol. 2001;19:54852.
Zhang XR, Mason H. Bean yellow dwarf virus replicons for high-level transgene
expression in transgenic plants and cell cultures. Biotechnol. Bioeng. 2006;93:
2719.
Zhao ZD, Tan L, Showalter AM, Lamport DTA, Kieliszewski MJ. Tomato LeAGP-1
arabinogalactan-protein puried from transgenic tobacco corroborates the Hyp
contiguity hypothesis. Plant J. 2002;31:43144.
Zhong JJ. Biochemical engineering of the production of plant-specic secondary
metabolites by cell suspension cultures. Adv. Biochem. Eng. Biotechnol. 2001;72:
1-26.
Zhong JJ, Fujiyama K, Seki T, Yoshida T. A quantitative-analysis of shear effects on cellsuspension and cell-culture of Perilla frutescens in bioreactors. Biotechnol. Bioeng.
1994;44:64954.
Zhong C, Yang S, Yuan YJ. Molecular responses of phospholipids of Taxus cuspidata
(Japanese yew) to hydrodynamic shear stress in bubble columns. Biotechnol. Appl.
Biochem. 2009;53:26575.
Ziyad-Mohamed MT, Scragg AH. Plant cell immobilization in alginate and polyurethane
foam. In: Walker JM, editor. Plant Cell and Tissue Culture, vol. 6. Clifton, New Jersey:
The Humana Press Inc; 1990. p. 52536.
Zuo JR, Niu QW, Chua NH. An estrogen receptor-based transactivator XVE mediates
highly inducible gene expression in transgenic plants. Plant J. 2000;24:26573.

Anda mungkin juga menyukai