You are on page 1of 15

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/51648778

Green factory: Plants as bioproduction


platforms for recombinant proteins

Article in Biotechnology advances September 2011


DOI: 10.1016/j.biotechadv.2011.08.020 Source: PubMed

CITATIONS READS

67 574

5 authors, including:

Jay Xu Maureen Dolan


Arkansas State University - Jonesboro Arkansas State University - Jonesboro
67 PUBLICATIONS 1,246 CITATIONS 27 PUBLICATIONS 524 CITATIONS

SEE PROFILE SEE PROFILE

Giuliana Medrano Pamela Weathers


The University of Tennessee Health Science Worcester Polytechnic Institute
23 PUBLICATIONS 306 CITATIONS 124 PUBLICATIONS 2,932 CITATIONS

SEE PROFILE SEE PROFILE

All in-text references underlined in blue are linked to publications on ResearchGate, Available from: Maureen Dolan
letting you access and read them immediately. Retrieved on: 12 September 2016
Biotechnology Advances 30 (2012) 11711184

Contents lists available at SciVerse ScienceDirect

Biotechnology Advances
journal homepage: www.elsevier.com/locate/biotechadv

Research review paper

Green factory: Plants as bioproduction platforms for recombinant proteins


Jianfeng Xu a, Maureen C. Dolan a, Giuliana Medrano a, Carole L. Cramer a, Pamela J. Weathers b,
a
Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, United States
b
Biology & Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA 01609, United States

a r t i c l e i n f o a b s t r a c t

Available online 7 September 2011 Molecular farming, long considered a promising strategy to produce valuable recombinant proteins not only
for human and veterinary medicine, but also for agriculture and industry, now has some commercially avail-
Keywords: able products. Various plant-based production platforms including whole-plants, aquatic plants, plant cell
Molecular farming suspensions, and plant tissues (hairy roots) have been compared in terms of their advantages and limits.
Plant-made recombinant proteins Effective recombinant strategies are summarized along with descriptions of scalable culture systems and
Transgenic plants
examples of commercial progress and success.
Plant cell culture
Microalgae
2011 Elsevier Inc. All rights reserved.
Moss
Hairy roots
Duckweed

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1172
2. Classes of recombinant proteins produced by plants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1172
2.1. Therapeutic proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1172
2.2. Industrial proteins/enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1174
2.3. Biopolymers. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1174
3. Plant expression platforms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1175
3.1. Whole plants stable expression systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1175
3.1.1. Leaf-based . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1175
3.1.2. Seed-based . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1175
3.2. Whole plants transient expression system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1176
3.3. In vitro culture systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1176
3.3.1. Plant cell suspensions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1177
3.3.2. Hairy roots . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1177
3.3.3. Moss . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1177
3.4. Aquatic plants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1178
3.4.1. Duckweed . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1178
3.4.2. Microalgae . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1178
4. Culture scale-up and bioreactors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1178
5. Commercialization status and outlook . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1179
5.1. Plant-made industrial proteins/enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1179
5.2. Plant-made therapeutic proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1179
5.3. Plant-made biopolymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1180
6. Conclusion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1180
Acknowledgments. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1181
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1181

Corresponding author at: Dept. Biology & Biotechnology, Worcester Polytechnic Institute, 100 Institute Rd, Worcester, MA 01609, United States. Fax: + 1 508 831 5936.
E-mail address: weathers@wpi.edu (P.J. Weathers).

0734-9750/$ see front matter 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.biotechadv.2011.08.020
1172 J. Xu et al. / Biotechnology Advances 30 (2012) 11711184

1. Introduction strengths and weaknesses and is often best suited for certain classes
of recombinant proteins based on the market, scale, cost, and up-
Plants possess exceptional biosynthetic capacity including the abil- stream and downstream processing constraints of the particular pro-
ity to use the sun (photosynthesis) and/or very simple media to sup- tein product. A multiplicity of plant species can serve as hosts for
port signicant biomass and protein accumulation. Their potential plant-based bioproduction that comprise platforms ranging from in
for low-cost production of high quality and bioactive recombinant vitro cell and plant tissue cultures to whole plants grown under
protein is well documented (Obembe et al., 2011; Paul and Ma, glass and in the eld. Access to such platform diversity makes it pos-
2011; Sharma and Sharma, 2009). Plants successfully perform the ma- sible to respond rapidly for expressing novel recombinant proteins,
jority of post-translational modications important for many complex enables customizing and meeting scale-up needs, and provides op-
eukaryotic proteins and provide tremendous exibility in bioproduc- portunity for oral-based delivery of proteins. This review highlights
tion platforms that differentially address production scale, cost, safety, the advantages and challenges associated with each type of plant pro-
and regulatory issues. Plant-based systems provide the spectrum of duction platform and production strategy, and where appropriate,
production capacity ranging from plant/algal cell bioreactor systems will comment on important issues related to regulation. It also high-
for lower volume, higher value product to eld-grown commodity lights progress toward product commercialization.
crops with potential for metric tons of recombinant protein at highly
competitive costs. Because plants cannot harbor the human and ani- 2. Classes of recombinant proteins produced by plants
mal pathogens-of-issue for mammalian cell-based production sys-
tems, they bring signicant advantage in increased safety for According to their functions and applications, plant-made recombi-
patients (Pogue et al., 2010; Xu et al., 2011a). These biosafety advan- nant proteins can be generally categorized into three classes: therapeu-
tages also impact commercial aspects; they reduce purication costs tic proteins, industrial proteins/enzymes and biopolymers. Examples
and minimize risks associated with potential production shut-downs, of each of these classes are highlighted in Table 2 and discussed briey
facility decontamination, and supply limitations leading to unmet in the following sections.
patient/customer demand. Although costs to purify proteins from
plant systems will be comparable to microbial or mammalian cell cul- 2.1. Therapeutic proteins
ture systems, lower up-front capitalization required to initiate com-
mercial production in plants and potential economies of scale Many proteins of mammalian origin have been expressed in
provide key advantages. Applications that directly use whole or mini- plants, yielding a product with full function. These include monoclo-
mally-processed plants or plant parts (e.g., seeds) are also being ex- nal antibodies (mAbs), vaccine antigens, therapeutic enzymes, blood
plored for industrial/bioenergy applications as well as therapeutics proteins, cytokines, growth factors and growth hormones (see ex-
and vaccines to further reduce recombinant protein costs (Boothe amples in Table 2) (Davies, 2010; De Muynck et al., 2010; Xu et al.,
et al., 2010; Hood, 2002; Howard et al., 2011). In many systems, boost- 2011a). Bioactivity of these proteins often requires protein folding,
ing protein yields remains a challenge for economic feasibility (Paul disulde bond formation, subunit assembly, proteolytic cleavage,
and Ma, 2011; Xu et al., 2011a) and key strategies used to boost prod- and/or glycosylation, highlighting the ability of plants to process
uct recovery in plant expression system are summarized in Table 1 complex human/mammalian proteins. Plant-made antibodies (Plan-
(see also Hood et al., 2011, in press; Rybicki, 2010). Plant-based pro- tibodies) have received considerable interest as they are made at
duction systems involving eld-grown GM plants require regulatory much lower cost in plants than in mammalian cells without the asso-
oversight and approvals that are unique to plants compared to other ciated risks of potentially harboring animal pathogens (De Muynck
bioproduction systems (Obembe et al., 2011); regulatory consider- et al., 2010). Plants in fact are currently recognized as the only viable
ations also impact production strategies and costs. production platform for the production of secretory antibodies
In contrast to other expression systems such as bacterial, mamma- (sIgAs) (Paul and Ma, 2011). These recombinant therapeutic pro-
lian cell and yeast, plant expression systems encompass diverse forms teins are produced using a multiplicity of plant-based platforms
including whole-plants, suspension cells, hairy roots, moss, duck- that range from cultures cells to eld crops with the most common
weed, microalgae, etc. (Fig. 1). Each of the platforms has its own being tobacco species with protein yields varying dramatically
from 0.01% of total soluble protein (TSP) (Mikschofsky et al., 2009)
and 0.1 g/L (Kwon et al., 2003), up to 25% of TSP (Vunsh et al.,
2007) and 247 mg/L (McDonald et al., 2005). Several recent reviews
Table 1
of plant-made therapeutic proteins are available (Davies, 2010;
Strategies used to boost recombinant protein yields in plants. Obembe et al., 2011; Paul and Ma, 2011; Rybicki, 2010; Sharma and
Sharma, 2009; Tremblay et al., 2010; Weathers et al., 2010; Xu
Production stages Strategies
et al., 2011a), so we will focus here on selective characteristics of
Transcription Develop strong promoters, double enhanced the plant-based systems permitting comparisons with bacterial, fun-
promoters, hybrid promoters
gal and mammalian cell systems.
Use inducible promoters
Engineer better enhancers, activators or repressors Plants successfully glycosylate proteins at the signature recogni-
Translation Optimize 5- and 3-UTR (untranslated region); tion motif (N-X-S/T) for N-linked glycosylation. However, subsequent
Design preferred genetic codon. processing in the Golgi to complex glycans differs from that found in
Post-translation Target nascent proteins to sub-cellular compartments mammalian cells. Thus, a notable challenge in using plants as hosts
such as ER
Co-express with protease inhibitor and protein
for production of glycosylated therapeutic proteins is that plant-
co-factor/subunit; co-express antibody with antigen specic xylose and -1,3-fucose sugars may be added with a poten-
Express as fusion to a highly expressed and stable tial to alter bioactivity or immunogenicity in humans (Doran, 2000;
peptide Gomord et al., 2010). Plants also do not naturally synthesize animal
In vitro cell/tissue culture Optimize medium composition
or human-specic sugars, such as -1,4-galactose residues or sialic
(for bioreactor system) Supplement protein-stabilizing agents
In situ remove expressed protein acid (Gomord et al., 2005; Gomord et al., 2010; Paccalet et al.,
Process development Select and/or improve bioreactor design 2007). Recent efforts involving RNAi strategies that knock down
(for bioreactor system) Select culture strategy (e.g. batch vs. fed-batch vs. fucosyl- and xylosyl-transferases in Nicotiana benthamiana, Arabidop-
continuous culture) sis and Physcomitrella patens and transgenic lines expressing a human
Evaluate secretion potential
or chimeric -1,4-galactosyltransferase have been used to engineer
J. Xu et al. / Biotechnology Advances 30 (2012) 11711184 1173

Fig. 1. Various plant cell expression platforms for the production of recombinant proteins.

more human-like glycosylation machinery in plants (Bakker et al., chimaeric human -1,4-galactosyltransferase resulted in antibodies
2006; Schahs et al., 2007; Strasser et al., 2008). Furthermore, Agro-in- with human-like N-glycans (Vezina et al., 2009). Although signicant
ltrated N. benthamiana (plants inltrated with Agrobacterium tume- research has been focused on humanizing the glycan of plant-made
faciens) producing human antibodies, when co-expressed with a glycoproteins, the rst human-injected therapeutic with greatest

Table 2
Examples of the three classes of recombinant proteins being produced using plants.

Classication Recombinant proteins Plant expression platform References

Therapeutic proteins Antibody: anti-West Nile virus mAb Hu-E16a, Nicotiana benthamiana Lai et al. (2010), Ma et al. (1998)
Guy's 13 SIgA (CaroRx)b
Vaccine: H5N1, H1N1c N. benthamiana Shoji et al. (2008, 2011)
Therapeutic enzyme: glucocerebrosidased Carrot suspension cells Aviezer et al. (2009a, 2009b)
Blood protein: human serum albumine Potato Sijmons et al. (1990)
Cytokine: interlukin-12 Tobacco hairy roots Liu et al. (2009)
Growth factor: human epidermal growth factor Tobacco tissues Parsons et al. (2010)
Growth hormone: human growth hormonef Tobacco chloroplast Staub et al. (2000)
Oral therapeutic: human intrinsic factorg Arabidopsis thaliana Fedosov et al. (2003)
Industrial enzymes Cellulase Corn Hood et al. (2007, 2011)
-glucuronidaseh Corn Kusnadi et al. (1998)
Trypsinh Corn Woodard et al. (2003)
Avidinh Corn Hood et al. (1997), Murray et al. (2002)
-Amylasei Corn, tobacco Pen et al. (1992)
Laccase Corn Hood et al. (2003)
Biopolymers Spider silk proteins Tobacco, potato, Arabidopsis Menassa et al. (2004), Scheller et al. (2001), Yang et al. (2005)
Elastin-like polypeptides Tobacco Conley et al. (2009)
Collagensj Tobacco, corn Ruggiero et al. (2000), Xu et al. (2011b)
Plant gum Tobacco suspension cells Xu et al. (2005)
a
Recent on commercial route.
b
Phase II clinical trial completed by Planet Biotechnology; approved for use in the EU, but not marketed.
c
Phase II and phase I clinical trials for H5N1 and H1N1, respectively, completed by Medicago with positive results.
d
Phase III clinical trial completed by Protalix; the rst plant-made therapeutic protein entering commercial sector; FDA approval pending.
e
First complex human protein expressed in plant.
f
Early demonstration of high-yield protein expression in plant chloroplast.
g
Phase II clinical trial completed by Cobento Biotech AS; Marketed in the EU.
h
Sigma Aldrich products come as research enzyme or drug.
i
Recently (Feb, 2011) commercialized by Syngenta.
j
Under preclinical development by Medicago and Meristem Therapeutics.
1174 J. Xu et al. / Biotechnology Advances 30 (2012) 11711184

clinical experience (Protalix's taliglucerase; discussed further below) conversion to produce biofuels such as ethanol are currently interesting
did not trigger signicant antibody production in patients (Aviezer et candidates for commercialization (Obembe et al., 2011). A major hin-
al., 2009a, 2009b). drance to using plant-made industrial proteins/enzymes is regulatory;
Plant-specic post-translational modications (PTMs) may pre- very large acreage of transgenic plants will be planted. This will likely
sent an opportunity for not only producing biosimilars, but also com- require deregulation by federal regulatory agencies for eld growth
plex recombinant proteins with enhanced function and efcacy (Faye of those transgenic plants (Horn et al., 2004) and also wider acceptance
and Gomord, 2010). To this end, continued advances in understand- by the general public of open eld cultivation of transgenic plants for
ing and manipulating post-translational modications in plants will protein production. Large scale production of GM biofuel corn,
be a key to the full adoption of plant-based therapeutic proteins. In which is a major commodity food/feed crop, remains controversial.
fact, the second generation of plant-made pharmaceutical proteins However, the regulatory route and public acceptance for crops expres-
is emerging at the research level whereby therapeutic targets are spe- sing this class of enzymes should be aided by the fact that these en-
cically engineered to enhance the production or utility of plant- zymes do not act on humans or animals (no substrate) and are
based therapeutics. The goal in these systems is to exploit the biopro- already widespread in the human diet (considered G.R.A.S, generally
duction capacity of plants to produce new therapeutic proteins that recognized as safe). On the other hand, when as projected, global food
integrate novel motifs or fusion to facilitate protein assembly, deliv- supplies become limited, then uses of a food/feed crop for non food/feed
ery, trafcking, protein stability, serum longevity, or protein solubility products will eventually meet with resistance and new production solu-
in either the production host or the target organism (e.g., Cramer tions will need to be developed.
et al., 2010; Phoolcharoen et al., 2011; Xu et al., 2007, 2010).
2.3. Biopolymers
2.2. Industrial proteins/enzymes
Differing from those abundant plant biopolymers such as cellulose
Industrial proteins are characterized by the fact that they are used and starch, recombinant biopolymers discussed here specically refer
in very large quantities and must therefore be produced at very low to those made of proteins such as spider silk proteins, elastin-like
cost (Horn et al., 2004). Transgenic plants provide a viable technology polypeptides (ELPs), collagens and plant gums (Table 2). The proper-
for producing industrial proteins, in particular enzymes, because of ties and recombinant expression of these biopolymers in plants were
low cost of agricultural production, stability of protein stored in spe- reviewed previously by Scheller and Conrad (2005). Of special inter-
cic organs such as seeds, ease and speed of scale-up as well as the est are the spider silk proteins (also called spidroins) that are modu-
possibility of using crude plant materials directly in industrial pro- lar brous proteins containing highly repetitive amino-acid
cesses (Hood, 2002). Some plant-made industrial proteins/enzymes sequences consisting largely of glycine and alanine (Hinman et al.,
that have received considerable interest are listed in Table 3; most of 2000; Scheller et al., 2001). Spider silk bers spun from these spi-
them are hydrolases, including glycosidases (e.g., cellulase, -amylase droins are regarded as one of nature's most extraordinary materials
and -glucuronidase (GUS)) and proteases (e.g., trypsin). ProdiGene with exceptional exibility, elasticity, and toughnessthree times as
Inc (formerly in College Station, TX) was the rst company to develop strong as Kevlar and ve times as strong as steel (Eisoldt et al.,
and commercialize plant-based recombinant proteins/enzymes with 2011). Plants offer a more efcient and cheaper production platform
GUS and Avidin being ProdiGene's rst two commercialized products than bacteria for production of recombinant spidroins. The expres-
(Basaran and Rodriguez-Cerezo, 2008; Hood, 2002; Sharma and sion of Nephila clavipes dragline spidroins encoded by synthetic
Sharma, 2009). ProdiGene, a little ahead of its time technically, is unfor- genes in transgenic tobacco and potato plants has resulted in accu-
tunately now out of business. mulation of recombinant silk proteins up to a level of 2% of TSP in
Corn seed is regarded as an ideal platform for the production of the ER (Scheller et al., 2001). In another report, accumulation of drag-
industrial proteins/enzymes because corn has the largest annual line spidroins in Arabidopsis reached 18% of TSP in seeds (Yang et al.,
grain yield and relatively high seed protein content (10%), thus offer- 2005). Currently, the challenge to the application of such biomaterials
ing the highest potential recombinant protein yields per hectare is the lack of appropriate techniques such as ber spinning to con-
(Christou et al., 2005). Advantages of a seed-based production system vert the raw material into intermediate products (Scheller et al.,
include easy protein storage and the ability to apply the product directly 2001). Another interesting biopolymer is the ELPs that are comprised
to industrial processes, minimizing handling and enzyme manipulation of the repetitive pentapeptide sequence (VGVPG), which mainly
and preparation (Boothe et al., 2010; Hood, 2002). Corn-produced en- serves as thermally responsive tags for the non-chromatographic pu-
zymes such as cellulases and hemicellulases involved in biomass rication of recombinant proteins (Conley et al., 2009). ELP tags were

Table 3
Comparisons of different expression systems.

Expression system Commercially viable species Time for productiona Scalability Regulatory compliance

Whole plants
Stable transgenic plants Corn, soy, safower, rice 36 months Unlimited eld culture Difcult
Transient plants Nicotiana sp., lettuce 27 days Greenhouse limited Moderate

In vitro cultured plant cells and species


Hairy roots Nicotiana sp. 1430 days 20,000 L Easy
Cell suspension culture Tobacco BY-2, carrot, rice 720 days 100,000 L Easy
Moss Physcomitrella patens 1430 days 200 L Easy

Aquatic plants
Duckweed (closed system) Lemna sp., spirodela sp. 2040 days 10,000 L Moderate
Microalgae Chlamydomonas reinhardtii
Open system 2040 days Limited by water surface area Difcult
Photobioreactor 1430 days 10,000 L Moderate
Conventional bioreactor 720 days 200 L Easy
a
The time required to accumulate maximum amounts of recombinant proteins in a culture system after planting or bioreactor inoculation.
J. Xu et al. / Biotechnology Advances 30 (2012) 11711184 1175

found to signicantly enhance the production yield of a range of dif- With an average of 100 chloroplasts per plant cell, the plastid pre-
ferent recombinant proteins in plant leaves (Conley et al., 2011; sents an alternative integration site which, in practice, enables thou-
Patel et al., 2007). Compared with therapeutic proteins and industrial sands of copies of a given transgene to be expressed (Chebolu and
enzymes that have shown high potential for commercialization, Daniell, 2007; Oey et al., 2009; Ruhlman et al., 2010). Chloroplastic
plant-made biopolymers still have a long way to go before they are transformation and expression of chloroplast-targeted genes have
cost effective to market. been reviewed in depth in a recent special issue on Chloroplast Bio-
technology in the Plant Biotechnology Journal (June 2011). To date,
3. Plant expression platforms over 100 recombinant proteins have been expressed through chloro-
plast transformation including industrial enzymes, antibodies, bio-
The rapid development of plant genetic engineering technologies pharmaceutical proteins and vaccine antigens as noted in a recent
has expanded the diversity of well-established plant-based biopro- review by Clarke et al. (2011). While the chloroplast does support
duction platforms for recombinant proteins (Paul and Ma, 2011). many of the important post-translational modications desired for
These platforms include transgenic plants (using both stable and expressing complex proteins, current plastid expression technology
transient expression), duckweed, microalgae, and in vitro culture sys- is limited to production of non-glycosylated products.
tems: cell suspensions, hairy roots and moss protonema (Fig. 1). Each Among the limitations of targeting recombinant protein expression
of the platforms has its own strengths and weaknesses, which are de- to the leaf is the nite shelf life of this tissue and the need to immediate-
scribed here. A generalized comparison of the time needed for pro- ly process upon harvest to ensure product stability and quality. Al-
duction, scalability and regulatory compliance of different platforms though plant leaves are advantageous in terms of biomass yield, this
is shown in Table 3. tissue is generally more subject to damage than seeds. There are many
cases of recombinant proteins in leaves having associated stability and
3.1. Whole plants stable expression systems accumulation issues. Product yields in eld-grown materials can be
highly variable due to environmental impacts (both biotic and abiotic
To drive expression of foreign genes, many technologies for stable stresses) leading to increased consideration of growth in more con-
integration of the transgene into the production plant have been de- trolled conditions (e.g., under plastic or in greenhouses), especially for
veloped and used over the years. The choices of 1) production host, pharmaceutical applications. In addition to associated instability of the
2) gene integration site (nuclear vs. plastid), 3) the cellular compart- protein itself, the harvested material has a limited shelf life and there-
ment for recombinant protein accumulation, and 4) the plant tissue fore, must be processed immediately after harvest. It should also be
targeted for product expression in establishing stable transgenic noted that at scale, regulatory-compliant disposal of transgenic bio-
plant lines will depend on a variety of considerations that include mass waste may have volume and cost implications.
the post-translational modications required for attaining functional
protein product, the stability of the expressed foreign protein in the
given plant host, desired expression levels of product and the down- 3.1.2. Seed-based
stream purication costs as well as the size and cost constraints of the Recombinant protein expression targeted to plant seeds can over-
product market (see also Hood et al., 2011). come limitations of protein stability and storage associated with leaf
tissue (Boothe et al., 2010; Lau and Sun, 2009; Ma et al., 2003). Several
3.1.1. Leaf-based cereal grains including rice, wheat, barley, soybean and maize are
Tobacco has emerged as the leading plant platform for leaf-based commonly used as expression hosts (Ramessar et al., 2008a). Proteins
recombinant protein expression (Tremblay et al., 2010). The rst expressed in seeds are generally protected from proteolytic degrada-
clinical trial of a plant-produced biopharmaceutical, secretory anti- tion, and storage upwards of three years at room temperature (longer
body variant of Guy's 13, was done with a product expressed in with cold storage) resulted in minimal loss of protein activity. The
eld grown tobacco leaves by Planet Biotechnology Inc. (Kaiser, high protein content of seeds, ranging from 7 to10%, has translated
2008; Ma et al., 1998). Tobacco is genetically well studied and easily into high expression for several seed-targeted proteins (e.g. Hood
manipulated, is classied as a non-food/non-feed crop, produces et al., 1997; Horvath et al., 2000; Woodard et al., 2003; Xie et al.,
high biomass (upwards of 300 tons per acre) and is one of the best 2008; Xue et al., 2003; Yang et al., 2006; Yang et al., 2008). Further-
studied platforms to date for expressing recombinant biopharma- more, maize seeds producing HIV neutralizing antibody 2 G12 (a po-
ceuticals. The breeding of low nicotine and low alkaloid tobacco va- tential candidate for a topical microbicide), were not only shown to
rieties has been used to overcome potential toxicity carryover into have cost-effective production, but also simple downstream purica-
recombinant protein products (Menassa et al., 2001). In addition tion processes, which are considered the most signicant costs associ-
to tobacco, many other leafy crops have been used for stable expres- ated in the production of biopharmaceuticals (Ramessar et al., 2008c).
sion of proteins including lettuce, alfalfa and clover. The use of crops The potential to use agronomically important food/feed commod-
like alfalfa has additional benets; it is a perennial that xes nitro- ity crops and other seed crops for non-food/feed GM-based applica-
gen and displays notable homogeneity of N-glycosylated recombi- tions such as biofuel enzymes or pharmaceuticals raises several
nant proteins (Fischer et al., 2004). regulatory issues for both regulatory agencies and consumers. Regu-
Both nuclear and plastid integration have been used when expressing latory compliance for eld growth (e.g., USDA APHIS/EPA) will en-
recombinant proteins in leaf tissue. The choice of gene integration within compass concepts of product segregation and stewardship. Crops
the plant cell is generally dictated by the post-translational requirements expressing therapeutics or vaccines will likely remain under regulato-
of the target protein. For glycosylated proteins, nuclear integration of the ry oversight for the life of the product and require containment pro-
transgene is needed to enable proper processing of the protein in the tocols and documentation at each step in the process. In contrast,
endomembrane system (Gomord et al., 2010). This strategy has been for industrial enzymes which are/will be produced at large scale, pro-
used in successfully expressing over 100 glycosylated proteins including ducers will likely seek deregulation. One of the regulatory consider-
antibodies (e.g. Johansen et al., 1999; Ma et al., 1995; Fiedler et al., ations for food-based grains is that while there is the need to
1997), vaccines (e.g. Pogrebnyak et al., 2005) cytokines (e.g. Wang et segregate the bioproduct source grain from the food/feed supply
al., 2008; Medrano et al., 2010), growth hormones and industrial en- chain, they do have a G.R.A.S. status from the US Food and Drug Ad-
zymes in plant leaves. Benchabane et al. (2008) and Sharma and ministration (FDA) which may reduce downstream hurdles linked
Sharma (2009) have recently reviewed expression of endomembrane- with pharmaceutical use of these plant-based products (Ramessar
targeted proteins in plants. et al., 2008b, 2009).
1176 J. Xu et al. / Biotechnology Advances 30 (2012) 11711184

While the rst plant-derived commercialized product was pro- recombinant proteins in plants occurs within a shorter time, typically
duced in maize (Hood et al., 1997; Witcher et al., 1998), oil-seeds 24 days post-inltration, with recoveries typically ranging from ~0.1
are emerging as a promising platform for recombinant protein pro- to 180 g/g fresh leaf depending on the gene of interest (Condori
duction due to their inherently low associated proteolytic activity et al., 2009; Cramer et al., 1999, 2010; Liu et al., 2008; Medrano
and simplied protein isolation via oil body separations. Among the et al., 2009, 2010). Recently, transient-expressed xylanase targeted
leading companies in this plant biotechnology sector is SemBioSys to the endomembrane system was reported with product recoveries
Genetics (Calgary, Canada), which has developed the oleosin-fusion as high as 1 mg/g fresh leaf (Llop-Tous et al., 2011). A direct compar-
platform, in which the target recombinant protein is produced as a fu- ison of stable versus transient expression of identical transgenes sug-
sion with oleosin and accumulates in safower oilbodies (www. gests yields of 4 to 20-fold higher for the transient systems (using
sembiosys.com/). Application of this technology has highlighted the de35S promoter) on a leaf fresh weight basis (Medrano et al., 2009).
development of an exceptionally low cost human insulin biosimilar Plant viral vectors also use the same Agro-inltration system to in-
for improved accessibility and global distribution of insulin in coun- troduce DNA copies into the plant cell. This transient expression process
tering the global diabetes epidemic. Over the past several years, a generally produces higher levels of recombinant protein with yields
number of other seed-based platforms have emerged, including Ven- reported to be as high as 5 mg/g of fresh leaf for GFP (Marillonnet
tria Biosciences and Meristem Therapeutics, which have invested in et al., 2004). However, recombinant protein expression by this process
eld-grown rice for the production of the human proteins lactoferrin takes up to 14 days, which can present issues for proteins prone to pro-
and lysozyme (www.ventria.com/). ORF Genetics Ltd., based in Ice- tease degradation and instability. Geneware (Kentucky BioProcessing
land, has targeted barley grain as the expression site for a number LLC; originally developed by Large Scale Biology Corp.), MagnIcon
of human cytokines and growth factors (www.orfgenetics.com/). (Icon Genetics) and Geminiviral expression systems (Arizona State Uni-
versity) are some examples of effective viral vector expression systems
3.2. Whole plants transient expression system (Gleba et al., 2005, 2007; Huang et al., 2009a; Mason et al., 2002).
Alpha-trichosanthin and human lysosomal acid lipase have been
Transient expression systems, mediated by recombinant viral or expressed using Geneware technology (Kumagai et al., 1993). MagnI-
plant binary vectors, are being increasingly used for the expression of con has been used to express hepatitis B core antigen (Huang et al.,
biopharmaceuticals in plants due to speed, high protein yields, and reg- 2006, 2008), Norwalk virus-like particles (Santi et al., 2008), and Yersi-
ulatory considerations. During this process, foreign genes are typically nia pestis antigens F1 and V (Santi et al., 2006). Geminivirus technology
introduced into leaf tissue of intact plants (generally non-transgenic) was used to produce both the heavy and light chain subunits of IgG mAb
by vacuum inltration of engineered Agrobacterium (containing 6D8 against Ebola virus GP1 (Wilson et al., 2000), hepatitis B core anti-
gene(s) of interest within T-DNA with or without additional virus-der- gen (HBc) and Norwalk virus capsid protein (Huang et al., 2009b). The
ived components). Recombinant protein production (based on extra- Launch vector system includes a combination of a plant viral vector ge-
chromosomal gene expression within the plant cell) is initiated in the nome and a binary plasmid of Agrobacterium in which several proteins
leaves within 24 h and continues for several days (Agro-mediated) to including inuenza targets have been expressed with recoveries of
several weeks (viral-mediated) depending on vector and protein. 60 g/g fresh leaf for recombinant H5HA-I (Musiychuk et al., 2007;
While chromosomal integration is possible, it occurs at considerably Shoji et al., 2009; Yusibov and Mamedov, 2010). Additional examples
lower frequency in comparison to the number of cells transiently and yield information of viral vector-mediated bioproduction are sum-
expressing the desired gene (Circelli et al., 2010; Sheludko, 2008). In marized elsewhere (Daniell et al., 2009; Sheludko, 2008). Examples of
contrast to stable transformation and when used as production systems, proteins in clinical development or commercialized were recently listed
these transgenic plants are grown only in contained environments, e.g., by Yusibov et al. (2011).
greenhouses. This system is widely used for assessing gene expression Recombinant protein production using transient expression is now
constructs in plants and to validate activity of new recombinant pro- being mobilized to large scale by industry. For example, the Fraunhofer
teins (Circelli et al., 2010; Daniell et al., 2009; Dhillon et al., 2009; Center for Molecular Biotechnology (Newark, DE) (http://www.
Lombardi et al., 2009; Pogue et al., 2010; Tremblay et al., 2010, fraunhofer.org) has developed a scalable, automated plant-based facto-
2011; Voinnet et al., 2003; Whaley et al., 2011). Biomass production ry using plant viral vector technology to efciently produce large
for transient systems typically involves non-transgenic plants (gen- amounts of pharmaceuticals within weeks. Medicago Inc. (Quebec, Can-
erally Nicotiana benthamiana or other Nicotiana species) often ada) (http://www.medicago.com) has a cGMP manufacturing facility
grown in contained environments, e.g., greenhouses, prior to inl- using a transient expression system to produce vaccines for Phase II
tration, which is a highly automated process at production scales. clinical trials. Icon Genetics (Bayer; Halle, Germany) (http://www.
In addition to rapid transgene assessment and production runs and icongenetics.com) also has a GMP manufacturing facility. Texas A&M
high product yields, these systems also provide the ability to easily (College Station, TX) and Kentucky BioProcessing LLC (Owensboro,
co-express several genes at the same time (e.g., Huang et al., 2010; KY) (http://www.kbpllc.com/) have constructed a GMP biomanufactur-
Medrano et al., 2009). However, transiently expressed products re- ing facility. Terrasphere (http://terraspheresystems.com/) has devel-
quire sizable greenhouse space and inltration infrastructure, imme- oped a high-density vertical hydroponic cultivation system that can
diate processing of harvested leaf biomass, and, if using the product be used year round to produce large quantities of transient plants and
in humans or animal, an additional purication step to remove endo- their products. The production of large quantities of recombinant pro-
toxins derived from the inltrated Agrobacterium. tein offered by transient plant expression systems, coupled with use
Transient expression is typically carried out in leaves of N. of current technology to increase yields and many technical promising
benthamiana, but also has been demonstrated to work in other plants solutions seem to favorably compare with mammalian or insect cell-
including potato, green pea, Arabidopsis, lettuce, and other Nicotiana based systems in quality, cost, and scale.
species such as N. debneyi, N. excelsior, N. exigua, N. maritime, and N.
simulans (Bhaskar et al., 2009; Green et al., 2009; Kim et al., 2009;
Negrouk et al., 2005; Sheludko et al., 2007; Sindarovska et al., 2008, 3.3. In vitro culture systems
2010). There are two basic approaches for plant transient expression
based on the mechanism by which the transgene is transferred into In vitro culture systems are characterized by the fact that plant
the plant cell: viral-mediated or plant binary vector-based (Fischer biomass is cultured in conned bioreactors under sterile conditions
et al., 1999; Matoba et al., 2011). In a transient expression system for large-scale production of recombinant proteins. Plant suspension
using conventional plant expression vectors, the accumulation of cells, hairy roots and moss fall into this category.
J. Xu et al. / Biotechnology Advances 30 (2012) 11711184 1177

3.3.1. Plant cell suspensions 3.3.2. Hairy roots


Like microorganisms, undifferentiated clusters of plant cells (callus) Hairy roots are generated by infection of plants with Agrobacterium
can be dispersed and propagated in a liquid medium to generate stable rhizogenes that harbors a large root-inducing (Ri) plasmid (Shanks
cell suspension cultures, which can retain the same production capacity and Morgan, 1999). Integration of T-DNA carried on the Ri plasmid
as whole plants. Increased concerns about regulatory compliance and into the plant genome results in differentiation and growth at the in-
product safety of mammalian systems recently have renewed interest in fection site of neoplastic roots, called hairy roots (Guillon et al.,
plant cell cultures as an alternative production platform for complex 2006b; Shanks and Morgan, 1999). These induced root tissues can be
pharmaceutical proteins (Huang and McDonald, 2009; Shih and Doran, grown indenitely in vitro and have become a viable alternative pro-
2009; Xu et al., 2011a). These systems provide cGMP-compatible produc- duction platform for heterologous proteins as well as other plant me-
tion environments more acceptable to the established pharmaceutical in- tabolites (Giri and Narasu, 2000; Guillon et al., 2006a).
dustry with added benets of complex protein processing compared to Similar to suspension cells, hairy roots can be axenically cultured
bacteria and yeasts, and increase safety compared to mammalian cell sys- in a controlled environment suitable for the production of high-value
tems which can harbor human pathogens. In fact, the rst plant-produced pharmaceutical proteins under cGMP requirements. In addition, the
commercial therapeutic protein for human infusion, glucocerebrosidase, possible extracellular secretion of expressed proteins from cultured
is being produced in carrot cells by Protalix Biotherapeutics, Inc. (Israel; hairy roots, or rhizosecretion (Gaume et al., 2003), offers a simplied
http://www.protalix.com), although proof-of-concept was originally method for the recovery of foreign proteins from an inexpensive and
established previously using tobacco plants (Cramer et al., 1999; Radin well-dened medium. The advantages of rapidly growing hairy roots
et al., 1999). over suspension cells include long-term genotype and phenotype sta-
Plant cell suspension culture combines the advantages of whole- bility, efcient productivity and the ability of hairy roots to grow on
plants with those of microbial cultures (reviewed in Hellwig et al., hormone-free medium. Since the rst plant recombinant protein, a
2004; Huang and McDonald, 2009; Xu et al., 2011a). Although plant full-length murine IgG1, was successfully produced in hairy root cul-
cell cultures do not share the perspectives of unlimited scalability of ture N10 years ago (Wongsamuth and Doran, 1997), nearly 20 recom-
eld-cultivated whole plants, culture of plant cells in a sterile environ- binant proteins, including reporter proteins (e.g. GUS and GFP) (Lee
ment allows for precise control over growth conditions, batch-to-batch et al., 2007; Medina-Bolivar and Cramer, 2004), enzymes (Gaume et
product consistency, and a production process aligned with current al., 2003; Woods et al., 2008), antibodies (Martinez et al., 2005;
good manufacturing practices (cGMP). The associated containment as- Sharp and Doran, 2001), antigens (Ko et al., 2006; Kumar et al.,
pects of both production and product reduces the regulatory burden 2006; Rukavtsova et al., 2007), growth factors (Komarnytsky et al.,
compared to eld-grown plants and provides a platform more consistent 2006; Parsons et al., 2010), and immunomodulators such as ricin-B
with established biopharmaceutical production systems potentially aid- (the non-toxic lectin subunit of ricin as antigen carrier) (Medina-
ing both regulatory and industry acceptance. The reduced regulatory Bolivar et al., 2003) and interleukin-12 (Liu et al., 2008; Liu et al.,
costs may partly outweigh the currently lower productivities of this pro- 2009) have been reported to be expressed by hairy roots with protein
duction platform along with its potentially higher capital costs. A wide yields up to 3.3% of TSP (Woods et al., 2008).
array of biologically active recombinant proteins have been successfully There are, however, challenges for large scale culture of hairy roots
expressed in plant cells, particularly in BY-2 (Bright Yellow-2) and NT-1 because they have a tangled, lamentous morphology and often possess
(Nicotiana tabacum-1). These strains are fast-growing, robust and readily unique physiological characteristics. Improved bioreactor designs, e.g.
undergo Agrobacterium-mediated transformation and cell cycle synchro- the mist reactor, originally developed by Weathers and Giles (1988),
nization (Hellwig et al., 2004; Ozawa and Takaiwa, 2010; Su and Lee, that offer low hydrodynamic stress, yet achieve high volumetric oxygen
2007; Tremblay et al., 2010). transfer are needed for culture of hairy roots (Choi et al., 2006; Kim and
One bottleneck in exploiting plant cell suspension culture for com- Yoo, 1993; Liu et al., 2009; Sivakumar et al., 2010). A Swiss biotechnol-
mercial purposes has been low productivity. Exceptions are expres- ogy company, ROOTec Bioactives Ltd (http://www.rootec.com/), is cur-
sion of human proteins such as 1-antitrypsin, interleukin-12 and rently focusing on commercialization of plant-derived products with
hGM-CSF in rice cells using a sucrose-inducible RAmy3D promoter hairy roots grown in a mist bioreactor. Hairy roots appear to be an at-
(Huang et al., 2001; McDonald et al., 2005; Trexler et al., 2005; Shin tractive in vitro expression system with several advantages compared
et al., 2003, 2010), where up to 247 mg/L was achieved (McDonald to eld-grown plants and suspension cultured cells. Future research
et al., 2005). However, the growth rates and characteristics of rice should be focused on establishing effective and economical bioreactors
cell lines are inferior to those of tobacco BY-2 and NT-1 cell lines for industrial production.
(Hellwig et al., 2004) and the viability of rice cells is signicantly de-
creased when cultivated in a sucrose-starvation medium (Huang and 3.3.3. Moss
McDonald, 2009). To attain a reasonable prot margin, the productiv- The moss, P. patens, with its fully sequenced genome (http://www.
ity of plant cell cultures needs to increase 1050-fold, and this re- cosmoss.org/) is also providing a promising platform for producing re-
quires a systematic strategy to maximize the efciency of all stages combinant products. Knockout strains lacking xylosylation and fuco-
of the production pipeline from gene expression to cell culture, pro- sylation activity have been produced so that recombinant proteins
cess development, and nally downstream protein purication can be humanized (Koprivova et al., 2004), and galactosylation has
(Weathers et al., 2010; Xu et al., 2011a). been obtained by adding human glactosyltransferase to the xylosyl
Although whole-plant produced pharmaceuticals do not yet account and fucosyltranferase locus (Huether et al., 2005). Using the moss
for a signicant portion of the preclinical and clinical pipeline, plant platform, a variety of different engineered proteins have now been
cells and tissues are emerging as a more compliant alternative factory produced including IgG1 and IgG4 antibodies, human VEGF, erythro-
(Xu et al., 2011a). Currently, two biotechnology companies, Protalix poietin, and factor H (Buttner-Mainik et al., 2011; Decker and Reski,
BioTherapeutics, Inc. (Israel; http://www.protalix.com) and Dow 2008). Secretion into the culture medium has also been achieved
AgroSciences (Indianapolis, IN; http://www.dowagro.com/) have fo- and found to be most effective when P. patens endogenous signal se-
cused on the development and commercialization of pharmaceutical quences are used instead of human sequences (Schaaf et al., 2005).
proteins expressed by proprietary plant cultures, named ProCellEx The haploid juvenile gametophyte (protonema) stage of the plant
and Concert Plant-Cell-Produced System, respectively. The commer- is morphologically similar to lamentous green algae and readily cul-
cial successes of these companies will undeniably dene a new era in tured in liquid media in photobioreactors. By controlling pH or am-
the biopharmaceutical industry that promises to provide a growth op- monium tartrate levels in the medium, the moss can be maintained
portunity for plant-produced products. indenitely in the protonema stage, thereby preventing development
1178 J. Xu et al. / Biotechnology Advances 30 (2012) 11711184

to the leafy mature gametophyte (Hohe et al., 2002). P. patens sus- 10 h), supports easy nuclear and chloroplast transformation, and
pensions also seem quite resistant to sheer stress; growth is not can be grown either photoautotrophically or with acetate as a carbon
inhibited even at 400500 rpm in stirred tank reactors (Hohe et al., source. A variety of recombinant proteins have been successfully
2002). Greenovation GmbH has now developed both process technol- expressed in microalgae as documented in a number of reviews
ogy and proprietary strains of P. patens for contract production of de- (Cadoret et al., 2008; Franklin and Mayeld, 2004, 2005; Leon-Ba-
sired proteins up to 200 L using tubular or disposable rocker bag nares et al., 2004; Mayeld et al., 2007; Potvin and Zhang, 2010;
bioreactors (http://www.greenovation.com/). Specht et al., 2010; Walker et al., 2005).
Both nuclear and chloroplast genomes of microalgae have been
3.4. Aquatic plants successfully transformed for expressing recombinant proteins. How-
ever, nuclear transformants generally failed to accumulate recombi-
3.4.1. Duckweed nant proteins to the levels observed in chloroplasts, most likely due
An aquatic higher plant, duckweed, is another promising biopro- to nuclear silencing mechanisms (Specht et al., 2010). Thus, only
duction platform for recombinant proteins. Duckweed is the common the chloroplast system is currently regarded as feasible for commer-
name for Lemnaceae, a monocot plant family consisting of four major cial production (Potvin and Zhang, 2010). A variety of functional re-
genera: Lemna, Spirodela, Wolfa and Wolfella. As the world's smal- combinant proteins including an antibody (Mayeld and Franklin,
lest, fastest-growing and simplest owering plant (Zhang et al., 2005), a vaccine (Streateld, 2006; Surzycki et al., 2009), a growth
2010), duckweed seems ideal for the production of recombinant pro- factor (Rasala et al., 2010), a blood protein (Manuell et al., 2007),
teins (Stomp, 2005; Yamamoto et al., 2001). It is safe, fast-growing and an industrial enzyme phytases (Yoon et al., 2011), have been suc-
(doubling time as short as 36 h), easy to grow and harvest, and has cessfully expressed in the chloroplast of C. reinhardtii. The disadvan-
a high protein content (up to 45% dry weight) (Stomp, 2005). Duck- tage of chloroplast transformation is that the expressed proteins
weed is capable of producing complex proteins that are not easily lack posttranslational modications such as glycosylation (Franklin
made by bacteria and yeast, or made only at great cost over a long pe- and Mayeld, 2005). Although problematic for some products, in an-
riod of time by mammalian cells. Moreover, duckweed is edible and tibody production, this can provide a benet because lack of glycosyl-
thus offers an attractive system for oral vaccines (Popov et al., 2006; ation prevents antibodies from activating the immune system (Dance,
Rival et al., 2008). 2010; Tran et al., 2009).
Similar to other higher plants, duckweed can be transformed Recombinant protein production with microalgae is still in its in-
using either biolistics or A. tumefaciens. Resulting transgenic lines fancy. The development of economically viable microalgal expression
are stable for both gene insertion and expression. More than 20 ther- systems is currently hindered by a lack of effective and consistent
apeutic proteins including plasminogen (Spencer et al., 2010), aproti- transformation methods for a wider variety of species, low (nuclear
nin (Rival et al., 2008), monoclonal antibody (Cox et al., 2006), avian expression) or inconsistent (chloroplast expression) recombinant
inuenza H5N1 hemagglutinin (Guo et al., 2009) and interferon 2 protein yields, as well as the lack of production systems optimized
(De Leede et al., 2008) have been produced in duckweeds with ex- for large-scale growth and harvesting under photoautotrophic condi-
pression levels up to 7% of TSP (Stomp, 2005). A reporter protein tions (Specht et al., 2010). Systematic and concerted research efforts
gene, GFP, expressed in Spirodela oligorrhiza reached a protein yield that are both biological- and engineering-based such as optimization
of more than 25% of TSP, making it among the highest expressing sys- of promoters, regulatory elements and codon usage as well as devel-
tems for nuclear transformation in a higher plant (Vunsh et al., 2007). opment of improved photobioreactor culture systems will be critical
Biolex, Inc., based in Pittsboro, North Carolina (www.biolex.com), to the success of the microalgal production platform.
is the major biotechnology company developing the duckweed (Lem-
na)-based expression (LEX) system for producing pharmaceutical 4. Culture scale-up and bioreactors
proteins. Currently, the leading product of Biolex is a controlled-re-
lease interferon 2b (Locteron) for the treatment of hepatitis C, While the scale of eld-grown plants can be enlarged almost in-
which offers tolerability and dosing advantages over PEGylated inter- denitely at low cost, scale-up of the elite genotype is the critical
ferons currently on the market. Two other products that are in pre- rst step involving in vitro cultured plant cell and tissue systems.
clinical development are a human plasmin (BLX-155) that is an Large scale culture of these in vitro cultures, especially to obtain
anticoagulant, and an anti-CD20 mAb (BLX-301) (Paul and Ma, small plantlets (micropropagation) is still a major challenge. There
2011). For the duckweed platform to be economically viable for in- are two main categories of bioreactors typically used to culture plants
dustrial production, several issues need to be addressed including im- cells, tissues, and organs: liquid-phase and gas-phase reactors (Kim
proved protein expression levels, human glycosylation, more et al., 2002). Liquid-phase reactors are the most commonly used,
genomic information on duckweed and duckweed reproduction, but delivery of adequate oxygen to submerged cells or tissues re-
and methods for scale-up to commercial levels in both closed and mains a challenge. Most plant cells and tissues are susceptible to
open systems (Stomp, 2005). shear stress so, unlike microbial cells, aggressive agitation is not par-
ticularly benecial. As a result, gas-phase reactors were also devel-
3.4.2. Microalgae oped to enhance O2 delivery, but with minimal shear. Since gasses
Microalgae also offer potential for large-scale and cost-effective are very insoluble in liquids, gas-phase reactors expose the biomass
production of recombinant proteins. They integrate the merits of mi- to air or a gas mixture, and nutrients are delivered as gas-infused
crobes including rapid growth and ease of culture with those of droplets. Hybrid versions of both types of reactor also exist wherein
higher plants in performing post-translational modication and pho- the headspace gas spans a large area of relatively shallow liquid to en-
tosynthesis (Franklin and Mayeld, 2005; Potvin and Zhang, 2010; hance gas exchange to the submerged cells or tissues. Because there
Specht et al., 2010). Use of transgenic microalgae for production of re- are a number of recent reviews on this topic (Ducos et al., 2010;
combinant proteins has lagged behind other microorganisms, such as Huang and McDonald, 2009; Weathers et al., 2010, 2011; Xu et al.,
bacteria and yeasts (Specht et al., 2010), but recently there has been a 2009), only a brief summary is provided here and the reader is direct-
surge of interest in microalgae as a viable bioproduction system ed to those for details.
(Rasala et al., 2010). The majority of current work is performed Bioreactors typically used for larger scale culture of plant cells and
with the well-characterized green unicellular alga, Chlamydomonas tissues include stirred tank, airlift, bubble column, temporary immer-
reinhardtii (Potvin and Zhang, 2010; Rosenberg et al., 2008; Rasala sion, balloon-type bubble, ebb and ow, mist reactor, and the rotating
et al., 2010). This alga has a rapid growth rate (doubling time of drum. Typical micropropagation uses a very labor- and cost-intensive
J. Xu et al. / Biotechnology Advances 30 (2012) 11711184 1179

process involving small culture boxes with semi-solid medium. De- enzyme, laccase (Hirai et al., 2008; Hood et al., 2003). For example,
sign and use of different reactor types and scale-up have been devel- an Arkansas-based small company, Innite Enzymes (Jonesboro,
oped for micropropagation and other plant production systems AR; http://www.inniteenzymes.com/index.htm) is engaged in de-
including cell suspensions, microalgae, hairy roots, moss protonema, veloping a cost-effective corn seed production system for cellulases
duckweed and other aquatic small plants, and even co-culture of mul- for cellulosic ethanol production. However, few products entered
tiple species. There are also many plant production systems being de- the marketplace until 2011 when a transgenic variety of corn expres-
veloped that instead use disposable (single use) bioreactors for sing -amylase was approved (deregulated for eld use) by the
culture of plant cells and tissues. These usually employ plastic bags USDA. This -amylase-producing crop was developed by Base-
of various designs including as a liner inside another vessel, a hanging l-based Syngenta and marketed as Enogen (http://www2.syngenta.
bag, the horizontal wave and wave and undertow bioreactors, the com/en/index.html). Unfortunately, it has sparked worries not only
slug bubble, the temporary immersion system (Box-In-Bag; RITA), from anti-GM organizations, but also from some biotechnology sup-
and a mist bag reactor. These newer disposable and issues regarding porters because of environmental and human health questions
their scale-up are described in greater detailed in recent reviews (Waltz, 2011).
(Eibl et al., 2010; Eibl and Eibl, 2008; Weathers et al., 2010, 2011).
5.2. Plant-made therapeutic proteins
5. Commercialization status and outlook
Because of the safety and economic concerns of using mammalian
Plants and their cells or organs are essentially a production factory cell cultures, plant-made therapeutic proteins are receiving renewed
for recombinant proteins. They provide both economic and safety ad- interest by pharmaceutical companies. In fact, several plant-made
vantages over conventional production systems. It was estimated that mammalian proteins such as human lactoferrin and lysozyme (Ven-
with 1% (w/w) of protein expression in corn on the dry weight base tria Bioscience), human aprotonin (ProdiGene), mammalian gastric
as well as 50% of protein recovery during purication, the cost of lipases (Meristem Therapeutics), and human intrinsic factor (Cobento
plant-produced proteins is 10 to 50- fold lower than that of micro- Biotechnology, Aarhus, Denmark) are now on the market (Obembe
bial systems, and up to 1000-fold lower than that of mammalian et al., 2011; Paul and Ma, 2011; Sharma and Sharma, 2009). However,
cell culture systems (Chen et al., 2005; Hood, 2002; Twyman et they are regarded as either nutraceutical or diagnostic reagents rather
al., 2003), implying the great potential of transgenic plants as an than pharmaceuticals. As of July 2011, there is no plant-made thera-
economically competent production platform. The molecular farm- peutic protein approved for human systemic use by the US FDA.
ing industry has grown rapidly in the past 15 years. While some Antibodies and vaccines are the two major classes of plant-made
plant-made industrial proteins/enzymes have been commercial- therapeutic proteins that are under commercial development. Planet
ized, many plant-made pharmaceuticals are in various develop- Biotechnology took the lead in developing and commercializing Plan-
mental stages. Examples of some successful plant-produced tibodies produced in tobacco; those being tested in clinical trials in-
products, either on the market or in commercial development, are clude CaroRX (for dental caries), DoxoRX (for side-effects of cancer
listed in recent reviews (Obembe et al., 2011; Paul and Ma, 2011; therapy), RhinoRX (for Rhinovirus prophylactic) and an IgG
Sharma and Sharma, 2009), or on the Molecular Farming website (ICAM1) (for common cold) (Obembe et al., 2011). In addition, a
last updated in 2010 (http://www.molecularfarming.com/PMPs- 2G12 IgG used as prophylactic treatment for HIV is in Phase I clinical
and-PMIPs.html). The major companies involved in research and trials by the Pharma-Planta Consortium (Paul and Ma, 2011), and a
development of plant-produced recombinant proteins include: Biolex plant-made scFv monoclonal antibody, used in downstream proces-
Therapeutics Inc. (Pittsboro, NC; http://www.biolex.com), BioStrategies sing of a hepatitis B vaccine, has been commercialized in Cuba
LC (Jonesboro, AR; http://www.biostrategies-lc.com), Dow AgroSciences (Pujol et al., 2005).
(Indianapolis, IN; http://www.dowagro.com), Fraunhofer Center for Mo- Subunit vaccine antigens produced in plants have received the most
lecular Biotechnology (Newark, DE; http://www.fraunhofer.org), Ken- attention in the molecular farming community. In 2006, Dow AgroS-
tucky BioProcessing LLC (Owensboro, KY; http://www.kbpllc.com/), ciences received the world's rst regulatory approval by the USDA for a
Medicago Inc. (Qubec, Canada; http://www2.medicargo.com/en), Meri- tobacco cell-based vaccine against Newcastle disease virus, which served
stem Therapeutics LLC (Cambridge, MA; http://www.meristem- as a landmark for production of therapeutic (veterinary) vaccines in
therapeutics.com), Protalix Biotherapeutics (Carmiel, Israel; http:// plants. One of the unique features of plant-produced vaccines is that
www.protalix.com), Planet Biotechnology (Hayward, CA; http://www. plants not only serve as the production bioreactor but also as the deliv-
planetbiotechnology.com), SemBioSys (Calgary, Canada; http://www. ery vehicle for oral vaccines (Tiwari et al., 2009; Wang and Coppel, 2008).
sembiosys.com) and Ventria Bioscience (Fort Collins, CO; http://www. Several oral vaccines have been subjected to Phase I clinical trials, e.g., a
ventria.com/). potato tuber-derived HBsAg (Hefferon, 2010). The challenges of develop-
ing oral vaccines lie in the difculty in dose standardization, concerns
5.1. Plant-made industrial proteins/enzymes linked to potential contamination of the food chain, and the low efcacy
of the oral route for immunization. However, the lower production and
Plant-made industrial proteins/enzymes have spearheaded com- delivery costs of these technologies may be essential for bringing these
mercialization over the other two classes of recombinant proteins; products to many areas of the world (Paul and Ma, 2011). The production
this progress is mainly credited to ProdiGene's pioneering work on of vaccines using a transient plant expression system has been developed
the development and commercialization of hydrolases including rapidly, somewhat due to the recent outbreak of avian and swine u,
GUS, avidin and trypsin (TrypZean) and aprotonin (AproliZean) which led to the development of seasonal and pandemic inuenza vac-
in the late 1990s (Basaran and Rodriguez-Cerezo, 2008; Hood, 2002; cines. Such vaccines are currently in pre-clinical and clinical trials spon-
Hood et al., 1997; Sharma and Sharma, 2009; Witcher et al., 1998). sored by Medicago Inc, The Texas Plant-Expressed Vaccine Consortium
All of these products are produced in transgenic corn and sold by Sig- and the Fraunhofer Center for Molecular Biotechnology. In addition, a re-
ma-Aldrich (St Louis, MO) (Sharma and Sharma, 2009). The commercial search group led by Charles J. Arntzen (a pioneer in plant-made edible
success of ProdiGene's products was followed by many efforts to de- vaccines) at Arizona State University is engaged in developing several
velop and commercialize other plant-made enzymes, in particular, plant-based vaccines including Hepatitis B surface antigen (Phase I in let-
biomass-degrading enzymes such as cellulase, hemicellulase, tuce; Phase II in potato), Vibrio cholera vaccine, Heat-labile toxin B subunit
xylanase and ligninase (Hood et al., 2007, 2011; Sticklen, 2007), of Escherichia coli and the capsid protein of Norwalk virus (all are Phase I
-amylase (Pen et al., 1992), as well as an oxido-reductase fungal in potato) (Obembe et al., 2011; Paul and Ma, 2011).
1180 J. Xu et al. / Biotechnology Advances 30 (2012) 11711184

Fig. 2. Comparison of benets and challenges associated with the three major types of plant production platforms. Because they are common to all plant platforms, the advantages
of plants over other production systems, e.g., eukaryotic protein processing, lack of human pathogens, scalability options, are not listed here.

In addition to antibodies and vaccines, there is special interest in other bodily tissues, is under preclinical development by Medicago
plant-produced therapeutic enzymes. The most encouraging example Inc. (produced in alfalfa) and Meristem Therapeutics (produced in to-
is a plant (carrot) cell-produced recombinant glucocerebrosidase bacco and corn) (Merle et al., 2002; Paul and Ma, 2011; Ruggiero
(prGCD) used as enzyme replacement therapy for the treatment of et al., 2000).
Gaucher disease (Shaaltiel et al., 2007). This prGCD was developed
by Protalix and will be marketed by Pzer with the commercial
name of taliglucerase alpha (Uplyso). It was awarded temporary 6. Conclusion
authorization for use in France in 2010, and thereby became the
rst plant cell-made human pharmaceutical to enter the commercial Plant-based bioproduction of proteins has evolved into a complex
market. Phases IIII clinical trials were accepted by the FDA and arena supporting diverse production platforms that offer advantages
nal approval is currently pending in the USA. Patient acceptance of in scale, cost, safety, and regulatory acceptability. A comparison of
taliglucerase alpha was enhanced by supply and safety concerns the advantages and remaining challenges of the main plant produc-
over Genzyme's CHO-based glucocerebrosidase, Cerezyme. Produc- tion platforms is summarized in Fig. 2. As the eld matures, there is
tion of Cerezyme was shut down several times in 2010 by the FDA greater differentiation between industrial and pharmaceutical appli-
due to viral contamination and follow-on production issues leading cation with the industrial enzymes targeting eld-based production
to drug shortages. Against this backdrop, the availability of Protalix's primarily in seed/grain crops driven by scale and cost. In contrast,
taliglucerase alpha highlighted the key advantages in safety and therapeutic proteins are moving toward greater containment and
cost of the plant-derived enzyme. In addition, commercial partner- consistency through bioreactor-based systems or transient expres-
ship with Pzer, the largest pharmaceutical company in the world, sion. It appears that plant-produced biologicals, long held as one of
suggests that plant-based bioproduction systems are being recog- the golden prospects of biotechnology, have nally come into their
nized as competitive in this arena. Other plant cell-produced enzymes own with commercialization of major plant-produced recombinant
in preclinical and clinical development by Protalix include a human proteins. With the exception of several plant-made enzymes that
acetylcholinesterase (PRX-105) for several therapeutic and prophy- were commercialized early, most plant-made pharmaceuticals only
lactic indications, -galactosidase (PRX-102) for the treatment of recently entered the marketplace. Specically, the human therapeu-
Fabry disease, and a pr-antiTNF, a biosimilar version of etanercept tic, taliglucerase alpha by Pzer (Protalix technology), was the rst
(Enbrel) for the treatment of rheumatoid arthritis. to enter the commercial market. Others at various stages of clinical
trials are anticipated to appear in the market over the next few
years. Signicant progress has been made in the engineering of re-
5.3. Plant-made biopolymers combinant proteins for production by in vitro plant cell and tissue
cultures. Advances in bioreactor design and implementation have
The commercial development of plant-made biopolymers lags far further enhanced this potential, especially for growing more com-
behind the above two classes of recombinant proteins. Collagen, a plex 3-D cultures like hairy roots. Many early challenges related
type of human protein that is brous and connects and supports to low protein yields and non-human glycosylation for therapeutic
J. Xu et al. / Biotechnology Advances 30 (2012) 11711184 1181

application have been overcome for select proteins and the eld Davies HM. Commercialization of whole-plant systems for biomanufacturing of protein
products: evolution and prospects. Plant Biotechnol J 2010;8:84561.
continues to develop innovative solutions to enhance the competi- De Leede LG, Humphries JE, Bechet AC, Van Hoogdalem EJ, Verrijk R, Spencer DG. Novel
tiveness and economic impact of plant-based systems. The green controlled-release Lemna-derived IFN-alpha2b (Locteron): pharmacokinetics,
phyto-pharm is thus, emerging as an alternative and important pharmacodynamics, and tolerability in a phase I clinical trial. J Interferon Cytokine
Res 2008;28:11322.
source of safe and cost effective therapeutics as well as industrial De Muynck B, Navarre C, Boutry M. Production of antibodies in plants: status after
enzymes. twenty years. Plant Biotechnol J 2010;8:52963.
Decker EL, Reski R. Current achievements in the production of complex biopharmaceu-
ticals with moss bioreactors. Bioprocress Biosyst Eng 2008;31:39.
Acknowledgments Dhillon T, Chiera J, Lindbo J, Finer J. Quantitative evaluation of six different viral sup-
pressors of silencing using image analysis of transient GFP expression. Plant Cell
Rep 2009;28:63947.
This work was supported in part by the Worcester Polytechnic In- Doran PM. Foreign protein production in plant tissue cultures. Curr Opin Biotechnol
stitute and the Plant Powered Production (P3) Center funds provided 2000;11:199204.
through an NSF RII Arkansas ASSET Initiative (AR EPSCoR) grant and Ducos JP, Terrier B, Courtois D. Disposable bioreactors for plant micropropagation and
mass plant cell culture. Adv Biochem Eng Biotechnol 2010;115:89-115.
the Arkansas Biosciences Institute, the major research component of Eibl R, Eibl D-. Design of bioreactors suitable for plant cell and tissue cultures. Phyto-
the Arkansas Tobacco Settlement Proceeds Act of 2000. chem Rev 2008;7:5938.
Eibl R, Kaiser S, Lombriser R, Eibl D. Disposable bioreactors: the current state-of-the-art
and recommended applications in biotechnology. Appl Microbiol Biotechnol
References 2010;86:419.
Eisoldt L, Smith A, Scheibel T. Decoding the secrets of spider silk. Mater Today 2011;14:
Aviezer D, Almon-Brill E, Shaaltiel Y, Galili G, Chertkoff R, Hashmueli S, et al. Novel en- 806.
zyme replacement therapy for Gaucher disease: ongoing Phase III clinical trial with Faye L, Gomord V. Success stories in molecular farming-a brief overview. Plant Biotech-
recombinant human glucocerebrosidase expressed in plant cells. Mol Genet Metab nol J 2010;8:5258.
2009a;96:S134. Fedosov SN, Laursen NB, Nexo E, Moestrup SK, Petersen TE, Jensen EO, et al. Human in-
Aviezer D, Brill-Almon E, Shaaltiel Y, Hashmueli S, Bartfeld D, Mizrachi S, et al. A plant- trinsic factor expressed in the plant Arabidopsis thaliana. Eur J Biochem 2003;270:
derived recombinant human glucocerebrosidase enzyme-a preclinical and phase I 33627.
investigation. PLoS One 2009b;4:e4792. Fiedler U, Philips J, Artsaenko O, Conrad U. Optimisation of scFv antibody production in
Bakker H, Rouwendal GJ, Karnoup AS, Florack DE, Stoopen GM, Helsper JP, et al. An an- transgenic plants. Immunotechnol 1997;3:20516.
tibody produced in tobacco expressing a hybrid beta-1,4-galactosyltransferase is Fischer R, Vaquero-Martin C, Sack M, Drossard J, Emans N, Commandeur U. Towards
essentially devoid of plant carbohydrate epitopes. Proc Natl Acad Sci U S A molecular farming in the future: transient protein expression in plants. Biotechnol
2006;103:757782. Appl Biochem 1999;30(Pt 2):1136.
Basaran P, Rodriguez-Cerezo E. An assessment of emerging molecular farming activi- Fischer R, Stoger E, Schillberg S, Christou P, Twyman RM. Plant-based production of
ties based on patent analysis (2002 similar to 2006). Biotechnol Bioprocress Eng biopharmaceuticals. Curr Opin Plant Biol 2004;7:1528.
2008;13:30412. Franklin SE, Mayeld SP. Prospects for molecular farming in the green alga Chlamydomonas
Benchabane M, Goulet C, Rivard D, Faye L, Gomord V, Michaud D. Preventing unin- reinhardtii. Curr Opin Plant Biol 2004;7:15965.
tended proteolysis in plant protein biofactories. Plant Biotechnol J 2008;6:63348. Franklin SE, Mayeld SP. Recent developments in the production of human therapeutic
Bhaskar PB, Venkateshwaran M, Wu L, Ane JM, Jiang J. Agrobacterium-mediated tran- proteins in eukaryotic algae. Expert Opin Biol Ther 2005;5:22535.
sient gene expression and silencing: a rapid tool for functional gene assay in pota- Gaume A, Komarnytsky S, Borisjuk N, Raskin I. Rhizosecretion of recombinant proteins
to. PLoS One 2009;4:e5812. from plant hairy roots. Plant Cell Rep 2003;21:118893.
Boothe J, Nykiforuk C, Shen Y, Zaplachinski S, Szarka S, Kuhlman P, et al. Seed-based ex- Giri A, Narasu ML. Transgenic hairy roots: recent trends and applications. Biotechnol
pression systems for plant molecular farming. Plant Biotechnol J 2010;8:588606. Adv 2000;18:1-22.
Buttner-Mainik A, Parsons J, Jerome H, Hartmann A, Lamer S, Schaaf A, et al. Production Gleba Y, Klimyuk V, Marillonnet S. Magnifection a new platform for expressing re-
of biologically active recombinant human factor H in Physcomitrella. Plant Biotech- combinant vaccines in plants. Vaccine 2005;23:20428.
nol J 2011;9:37383. Gleba Y, Klimyuk V, Marillonnet S. Viral vectors for the expression of proteins in plants.
Cadoret JP, Bardor M, Lerouge P, Cabigliera M, Henriquez V, Carlier A. Microalgae as cell fac- Curr Opin Biotechnol 2007;18:13441.
tories producing recombinant commercial proteins. Med Sci (Paris) 2008;24:37582. Gomord W, Chamberlain P, Jefferis R, Faye L. Biopharmaceutical production in plants:
Chebolu S, Daniell H. Stable expression of Gal/GalNAc lectin of Entamoeba histolytica in problems, solutions and opportunities. Trends Biotechnol 2005;23:55965.
transgenic chloroplasts and immunogenicity in mice towards vaccine develop- Gomord V, Fitchette AC, Menu-Bouaouiche L, Saint-Jore-Dupas C, Plasson C, Michaud D,
ment for amoebiasis. Plant Biotechnol J 2007;5:2309. et al. Plant-specic glycosylation patterns in the context of therapeutic protein
Chen M, Liu XW, Wang ZK, Song J, Qi QS, Wang PG. Modication of plant N-glycans production. Plant Biotechnol J 2010;8:56487.
processing: the future of producing therapeutic protein by transgenic plants. Green BJ, Fujiki M, Mett V, Kaczmarczyk J, Shamloul M, Musiychuk K, et al. Transient
Med Res Rev 2005;25:34360. protein expression in three Pisum sativum (green pea) varieties. Biotechnol J
Choi YE, Kim YS, Paek KY. Types and designs of bioreactors for hairy root culture. In: 2009;4:2307.
Dutta Gupta S, Ibaraki Y, editors. Plant tissue culture engineering focus on bio- Guillon S, Tremouillaux-Guiller J, Pati PK, Rideau M, Gantet P. Hairy root research: re-
technology. Dordrecht, The Netherlands: Springer; 2006. p. 16172. cent scenario and exciting prospects commentary. Curr Opin Plant Biol
Christou P, Stoger E, Twyman RM. Monocot expression system for molecular farming. 2006a;9:3416.
In: Fischer R, Schillbe S, editors. Molecular farming: plant-made pharmaceuticals Guillon S, Tremouillaux-Guiller J, Pati PK, Rideau M, Gantet P. Harnessing the potential
and technical proteins. Weinheim: John Wiley and Sons; 2005. p. 5567. of hairy roots: dawn of a new era. Trends Biotechnol 2006b;24:4039.
Circelli P, Donini M, Villani ME, Benvenuto E, Marusic C. Efcient Agrobacterium-based Guo X, Bublot M, Pritchard N, Dickey L, Thomas C, Swayne DE. Lemna (duckweed)
transient expression system for the production of biopharmaceuticals in plants. expressed hemagglutinin from avian inuenza H5N1 protects chickens against
Bioeng Bugs 2010;1:2214. H5N1 high pathogenicity avian inuenza virus challenge. Abstracts of the 7th
Clarke JL, Daniell H, Nugent JM. Chloroplast biotechnology, genomics and evolution: International Symposium on Avian Inuenza, 2009; Athens, Georgia; 2009.
current status, challenges and future directions. Plant Mol Biol 2011;76:2079. p. 62.
Condori J, Medrano G, Sivakumar G, Nair V, Cramer C, Medina-Bolivar F. Functional Hefferon K. Clinical trials fuel the promise of plant-derived vaccines. Am J Clin Med
characterization of a stilbene synthase gene using a transient expression system 2010;7:307.
in planta. Plant Cell Rep 2009;28:58999. Hellwig S, Drossard J, Twyman RM, Fischer R. Plant cell cultures for the production of
Conley AJ, Joensuu JJ, Jevnikar AM, Menassa R, Brandle JE. Optimization of elastin-like recombinant proteins. Nat Biotechnol 2004;22:141522.
polypeptide fusions for expression and purication of recombinant proteins in Hinman MB, Jones JA, Lewis RV. Synthetic spider silk: a modular ber. Trends Biotech-
plants. Biotechnol Bioeng 2009;103:56273. nol 2000;18:3749.
Conley AJ, Joensuu JJ, Richman A, Menassa R. Protein body-inducing fusions for high- Hirai H, Kashima Y, Hayashi K, Sugiura T, Yamagishi K, Kawagishi H, et al. Efcient ex-
level production and purication of recombinant proteins in plants. Plant Biotech- pression of laccase gene from white-rot fungus Schizophyllum commune in a trans-
nol J 2011;9:41933. genic tobacco plant. FEMS Microbiol Lett 2008;286:1305.
Cox KM, Sterling JD, Regan JT, Gasdaska JR, Frantz KK, Peele CG, et al. Glycan optimiza- Hohe A, Decker EL, Gorr G, Schween G, Reski R. Tight control of growth and cell differ-
tion of a human monoclonal antibody in the aquatic plant Lemna minor. Nat Bio- entiation in photoautotrophically growing moss (Physcomitrella patens) bioreactor
technol 2006;24:15917. cultures. Plant Cell Rep 2002;20:113540.
Cramer CL, Boothe JG, Oishi KK. Transgenic plants for therapeutic proteins: linking up- Hood EE. From green plants to industrial enzymes. Enzyme Microb Technol 2002;30:
stream and downstream strategies. Curr Top Microbiol Immunol 1999;240:95-118. 27983.
Cramer CL, Dolan MC, Reidy MJ. 2010. Methods of delivery of molecules to cells using Hood EE, Witcher DR, Maddock S, Meyer T, Baszczynski C, Bailey M, et al. Commercial
ricin subunit and compositions relating to same. United State Patent Application, production of avidin from transgenic maize: characterization of transformant, pro-
No. 20100240597. duction, processing, extraction and purication. Mol Breed 1997;3:291306.
Dance A. From pond scum to pharmacy shelf. Nat Med 2010;16:1469. Hood EE, Bailey MR, Beifuss K, Magallanes-Lundback M, Horn ME, Callaway E, et al. Cri-
Daniell H, Singh ND, Mason H, Streateld SJ. Plant-made vaccine antigens and biophar- teria for high-level expression of a fungal laccase gene in transgenic maize. Plant
maceuticals. Trends Plant Sci 2009;14:66979. Biotechnol J 2003;1:12940.
1182 J. Xu et al. / Biotechnology Advances 30 (2012) 11711184

Hood EE, Love R, Lane J, Bray J, Clough R, Pappu K, et al. Subcellular targeting is a key Ma JK, Hiatt A, Hein MB, Vine ND, Wang F, Stabila P, et al. Generation and assembly of
condition for high-level accumulation of cellulase protein in transgenic maize secretory antibodies in plants. Science 1995;268:7159.
seed. Plant Biotechnol J 2007;5:70919. Ma JK, Hikmat BY, Wycoff K, Vine ND, Chargelegue D, Yu L, et al. Characterization of a
Hood EE, Devaiah SP, Fake G, Egelkrout E, Teoh KT, Requesens DV, et al. Manipulating recombinant plant monoclonal secretory antibody and preventive immunotherapy
corn germplasm to increase recombinant protein accumulation. Plant Biotechnol in humans. Nat Med 1998;4:6016.
J 2011. doi:10.1111/j.1467-7652.2011.00627.x. Ma JK, Drake PMW, Christou P. The production of recombinant pharmaceutical pro-
Hood EE, Cramer CL, Medrano G, Xu J. Protein targeting: Strategic planning for optimiz- teins in plants. Nat Rev Genets 2003;4:794805.
ing protein products through plant biotechnology. In: Altman A, Hasagawa PM, ed- Manuell AL, Beligni MV, Elder JH, Siefker DT, Tran M, Weber A, et al. Robust expression
itors. Plant Biotechnology and Agriculture: Prospects for the 21st Century. Elsevier; of a bioactive mammalian protein in Chlamydomonas chloroplast. Plant Biotechnol
in press. doi: 10.1016/B978-0-12-381466-1.00003-1. J 2007;5:40212.
Horn ME, Woodard SL, Howard JA. Plant molecular farming: systems and products. Marillonnet S, Giritch A, Gils M, Kandzia R, Klimyuk V, Gleba Y. In planta engineering of
Plant Cell Rep 2004;22:71120. viral RNA replicons: efcient assembly by recombination of DNA modules deliv-
Horvath H, Huang J, Wong O, Kohl E, Okita T, Kannangara CG, et al. The production of recom- ered by Agrobacterium. Proc Natl Acad Sci U S A 2004;101:68527.
binant proteins in transgenic barley grains. Proc Natl Acad Sci U S A 2000;97:19149. Martinez C, Petruccelli S, Giulietti AMA, Alvarez MA. Expression of the antibody 14D9
Howard JA, Nikolov Z, Hood EE. Enzyme production systems for biomass conversion. in Nicotiana tabacum hairy roots. Electron J Biotechnol 2005;8:1706.
In: Hood EE, Nelson P, Powell R, editors. Plant Biomass Conversion. Ames, IA: Mason HS, Palmer KE, Hefferon KL, Mor TS, Arntzen C. 2002. Gemini virus vectors for
Wiley Press; 2011. p. 22753. gene expression in plants. United State Patent, No. 6392121
Huang TK, McDonald KA. Bioreactor engineering for recombinant protein production in Matoba N, Davis KR, Palmer KE. Recombinant protein expression in Nicotiana. Methods
plant cell suspension cultures. Biochem Eng J 2009;45:16884. Mol Biol 2011;701:199219.
Huang J, Sutliff TD, Wu L, Nandi S, Benge K, Terashima M, et al. Expression and puri- Mayeld SP, Franklin SE. Expression of human antibodies in eukaryotic micro-algae.
cation of functional human alpha-1-Antitrypsin from cultured plant cells. Biotech- Vaccine 2005;23:182832.
nol Prog 2001;17:12633. Mayeld SP, Manuell AL, Chen S, Wu J, Tran M, Siefker D, et al. Chlamydomonas rein-
Huang Z, Santi L, LePore K, Kilbourne J, Arntzen CJ, Mason HS. Rapid, high-level produc- hardtii chloroplasts as protein factories. Curr Opin Biotechnol 2007;18:12633.
tion of hepatitis B core antigen in plant leaf and its immunogenicity in mice. Vac- McDonald KA, Hong LM, Trombly DM, Xie Q, Jackman AP. Production of human
cine 2006;24:250613. alpha-1-antitrypsin from transgenic rice cell culture in a membrane bioreactor.
Huang Z, LePore K, Elkin G, Thanavala Y, Mason HS. High yield rapid production of hep- Biotechnol Prog 2005;21:72834.
atitis B surface antigen in plant leaf by a viral expression system. Plant Biotechnol J Medina-Bolivar F, Cramer C. Production of recombinant proteins by hairy roots cul-
2008;6:2029. tured in plastic sleeve bioreactors. Methods Mol Biol 2004;267:35163.
Huang C, Xie Y, Zhou X. Efcient virus-induced gene silencing in plants using a modi- Medina-Bolivar F, Wright R, Funk V, Sentz D, Barroso L, Wilkins TD, et al. A non-toxic lectin
ed geminivirus DNA1 component. Plant Biotechnol J 2009a;7:25465. for antigen delivery of plant-based mucosal vaccines. Vaccine 2003;21:997-1005.
Huang Z, Chen Q, Hjelm B, Arntzen C, Mason H. A DNA replicon system for rapid high-level Medrano G, Reidy MJ, Liu J, Ayala J, Dolan MC, Cramer CL. Rapid system for evaluating
production of virus-like particles in plants. Biotechnol Bioeng 2009b;103:70614. bioproduction capacity of complex pharmaceutical proteins in plants. Methods
Huang Z, Phoolcharoen W, Lai H, Piensook K, Cardineau G, et al. High-level rapid pro- Mol Biol 2009;483:5167.
duction of full-size monoclonal antibodies in plants by a single-vector DNA repli- Medrano G, Dolan MC, Stephens NT, McMickle A, Erf G, Radin D, et al. Efcient plant-
con system. Biotechnol Bioeng 2010;106:9-17. based production of chicken interleukin-12 yields a strong immunostimulatory cy-
Huether CM, Lienhart O, Baur A, Stemmer C, Gorr G, Reski R, et al. Glyco-engineering of tokine. J Interferon Cytokine Res 2010;30:14354.
moss lacking plant-specic sugar residues. Plant Biol 2005;7:2929. Menassa R, Hong Z, Karatzas CN, Lazaris A, Richman A, Brandle J. Spider dragline silk
Johansen FE, Natvig Norderhaug I, Roe M, Sandlie I, Brandtzaeg P. Recombinant expres- proteins in transgenic tobacco leaves: accumulation and eld production. Plant
sion of polymeric IgA: incorporation of J chain and secretory component of human Biotechnol J 2004;2:4318.
origin. Eur J Immunol 1999;;29:17018. Menassa R, Nguyen V, Jevnikar AN, Brandle JE. A self-contained system for the eld produc-
Kaiser J. Is the drought over for pharming? Science 2008;320:4735. tion of plant recombinant human interleukin-10. Mol Breed 2001;8:17785.
Kim YH, Yoo YJ. Development of a bioreactor for high-density culture of hairy roots. Merle C, Perret S, Lacour T, Jonval V, Hudaverdian S, Garrone R, et al. Hydroxylated
Biotechnol Techniques 1993;7:85962. human homotrimeric collagen I in Agrobacterium tumefaciens-mediated transient
Kim Y, Wyslouzil BE, Weathers PJ. Invited review: secondary metabolism of hairy root expression and in transgenic tobacco plant. FEBS Lett 2002;515:1148.
cultures in bioreactors. In Vitro Cell Dev Biol Plant 2002;38:1-10. Mikschofsky H, Schirrmeier H, Keil GM, Lange B, Polowick PL, Keller W, et al. Pea-der-
Kim MJ, Baek K, Park CM. Optimization of conditions for transient Agrobacterium- ived vaccines demonstrate high immunogenicity and protection in rabbits against
mediated gene expression assays in Arabidopsis. Plant Cell Rep 2009;28:115967. rabbit haemorrhagic disease virus. Plant Biotechnol J 2009;7:53749.
Ko S, Liu JR, Yamakawa T, Matsumoto Y. Expression of the protective antigen (SpaA) in Murray C, Sutherland PW, Phung MM, Lester MT, Marshall RK, Christeller JT. Expres-
transgenic hairy roots of tobacco. Plant Mol Biol Rep 2006;24:251. sion of biotin-binding proteins, avidin and streptavidin, in plant tissues using
Komarnytsky S, Borisjuk N, Yakoby N, Garvey A, Raskin I. Cosecretion of protease inhib- plant vacuolar targeting sequences. Transgenic Res 2002;11:199214.
itor stabilizes antibodies produced by plant roots. Plant Physiol 2006;141:118593. Musiychuk K, Stephenson N, Bi H, Farrance CE, Orozovic G, Brodelius M, et al. A launch
Koprivova A, Stemmer C, Altmann F, Hoffmann A, Kopriva S, Gorr G, et al. Targeted vector for the production of vaccine antigens in plants. Inuenza Other Respi Vi-
knockouts of Physcomitrella lacking plantspecic immunogenic N-glycans. Plant ruses 2007;1:1925.
Biotechnol J 2004;2:51723. Negrouk V, Eisner G, Lee H, Han K, Taylor D, Wong HC. Highly efcient transient expression
Kumagai MH, Turpen TH, Weinzettl N, della-Cioppa G, Turpen AM, Donson J, et al. of functional recombinant antibodies in lettuce. Plant Sci 2005;169:4338.
Rapid, high-level expression of biologically active alpha-trichosanthin in trans- Obembe OO, Popoola JO, Leelavathi S, Reddy SV. Advances in plant molecular farming.
fected plants by an RNA viral vector. Proc Natl Acad Sci U S A 1993;90:42730. Biotechnol Adv 2011;29:21022.
Kumar GBS, Ganapathi TR, Srinivas L, Revathi CJ, Bapat VA. Expression of hepatitis B Oey M, Lohse M, Kreikemeyer B, Bock R. Exhaustion of the chloroplast protein synthe-
surface antigen in potato hairy roots. Plant Sci 2006;170:91825. sis capacity by massive expression of a highly stable protein antibiotic. Plant J
Kusnadi AR, Evangelista RL, Hood EE, Howard JA, Nikolov ZL. Processing of transgenic 2009;57:43645.
corn seed and its effect on the recovery of recombinant beta-glucuronidase. Bio- Ozawa K, Takaiwa F. Highly efcient Agrobacterium-mediated transformation of sus-
technol Bioeng 1998;60:4452. pension-cultured cell clusters of rice (Oryza sativa L.). Plant Sci 2010;179:3337.
Kwon TH, Seo JE, Kim J, Lee JH, Jang YS, Yang MS. Expression and secretion of the het- Paccalet T, Bardor M, Rihouey C, Delmas F, Chevalier C, D'Aoust MA, et al. Engineering
erodimeric protein interleukin-12 in plant cell suspension culture. Biotechnol of a sialic acid synthesis pathway in transgenic plants by expression of bacterial
Bioeng 2003;81:8705. Neu5Ac-synthesizing enzymes. Plant Biotechnol J 2007;5:1625.
Lai HF, Engle M, Fuchs A, Keller T, Johnson S, Gorlatov S, et al. Monoclonal antibody pro- Parsons J, Wirth S, Dominguez M, Bravo-Almonacid F, Giulietti AM, Rodriguez Talou J.
duced in plants efciently treats West Nile virus infection in mice. Proc Natl Acad Production of human epidermal growth factor (hEGF) by in vitro cultures of Nico-
Sci U S A 2010;107:241924. tiana tabacum: effect of tissue differentiation and sodium nitroprusside addition.
Lau OS, Sun SSM. Plant seeds as bioreactors for recombinant protein production. Bio- Int J Biotechnol Biochem 2010;6:1318.
technol Adv 2009;27:101522. Patel J, Zhu H, Menassa R, Gyenis L, Richman A, Brandle J. Elastin-like polypeptide fu-
Lee KT, Chen SC, Chiang BL, Yamakawa T. Heat-inducible production of beta-glucuronidase sions enhance the accumulation of recombinant proteins in tobacco leaves. Trans-
in tobacco hairy root cultures. Appl Microbiol Biotechnol 2007;73:104753. genic Res 2007;16:23949.
Leon-Banares R, Gonzalez-Ballester D, Galvan A, Fernandez E. Transgenic microalgae as Paul M, Ma JKC. Plant-made pharmaceuticals: leading products and production plat-
green cell-factories. Trends Biotechnol 2004;22:4552. forms. Biotechnol Appl Biochem 2011;58:5867.
Liu J, Dolan M, Reidy M, Cramer C. Expression of bioactive single-chain murine IL-12 in Pen J, Molendijk L, Quax WJ, Sijmons PC, van Ooyen AJ, van den Elzen PJ, et al. Produc-
transgenic plants. J Interferon Cytokine Res 2008;28:38192. tion of active Bacillus licheniformis alpha-amylase in tobacco and its application in
Liu CZ, Towler MJ, Medrano G, Cramer CL, Weathers PJ. Production of mouse interleu- starch liquefaction. Nat Biotechnol 1992;10:2926.
kin-12 is greater in tobacco hairy roots grown in a mist reactor than in an airlift re- Phoolcharoen W, Bhoo SH, Lai H, Ma J, Arntzen CJ, Chen Q, et al. Expression of an im-
actor. Biotechnol Bioeng 2009;102:107486. munogenic Ebola immune complex in Nicotiana benthamiana. Plant Biotechnol J
Llop-Tous I, Ortiz M, Torrent M, Ludevid MD. The expression of a xylanase targeted to 2011. doi:10.1111/j.1467-7652.2011.00593.x.
ER-protein bodies provides a simple strategy to produce active insoluble enzyme Pogrebnyak N, Golovkin M, Andrianov V, Spitsin S, Smirnov Y, Egolf R, et al. Severe
polymers in tobacco plants. PLoS One 2011;6:e19474. Acute respiratory syndrome (SARS) S protein production in plants: development
Lombardi R, Circelli P, Villani ME, Buriani G, Nardi L, Coppola V, et al. High-level HIV-1 of recombinant vaccine. Proc Natl Acad Sci USA 2005;102:90627.
Nef transient expression in Nicotiana benthamiana using the P19 gene silencing Pogue GP, Vojdani F, Palmer KE, Hiatt E, Hume S, Phelps J, et al. Production of pharma-
suppressor protein of Artichoke Mottled Crinckle Virus. BMC Biotechnol 2009;9: ceutical-grade recombinant aprotinin and a monoclonal antibody product using
96. plant-based transient expression systems. Plant Biotechnol J 2010;8:63854.
J. Xu et al. / Biotechnology Advances 30 (2012) 11711184 1183

Popov SV, Golovchenko VV, Ovodova RG, Smirnov VV, Khramova DS, Popova GY, et al. Sijmons PC, Dekker BM, Schrammeijer B, Verwoerd TC, van den Elzen PJ, Hoekema A.
Characterisation of the oral adjuvant effect of lemnan, a pectic polysaccharide of Production of correctly processed human serum albumin in transgenic plants.
Lemna minor L. Vaccine 2006;24:54139. Nat Biotechnol 1990;8:21721.
Potvin G, Zhang ZS. Strategies for high-level recombinant protein expression in trans- Sindarovska YR, Sheludko YV, Gerasymenko IM, Bannikova MA, Kuchuk NV. Purica-
genic microalgae: a review. Biotechnol Adv 2010;28:9108. tion of recombinant GFP produced by Agrobacterium-mediated transient expres-
Pujol M, Ramirez NI, Ayala M, Gavilondo JV, Valdes R, Rodriguez M, et al. An integral sion in Nicotiana excelsior. Tsitol Genet 2008;42:1620.
approach towards a practical application for a plant-made monoclonal antibody Sindarovska YR, Gerasymenko IM, Sheludko YV, Olevinskaya ZM, Spivak NY, Kuchuk
in vaccine purication. Vaccine 2005;23:18337. NV. Production of human interferon alfa 2b in plants of Nicotiana excelsior by Agro-
Radin DN, Cramer, CL, Oishi KK, Weissenborn DL. 1999. Production of Human Lysosom- bacterium-mediated transient expression. Tsitol Genet 2010;44:604.
al Proteins in Plant-based Expression Systems. United State Patent. No. 5929304; Sivakumar G, Liu CZ, Towler MJ, Weathers PJ. Biomass production of hairy roots of Artemisia
European Patent. No. 0865499. annua and Arachis hypogaea in a scaled-up mist bioreactor. Biotechnol Bioeng 2010;107:
Ramessar K, Capell T, Christou P. Molecular pharming in cereal crops. Phytochem Rev 80213.
2008a;7:57992. Specht E, Miyake-Stoner S, Mayeld S. Micro-algae come of age as a platform for re-
Ramessar K, Sabalzaa M, Capell T, Christou P. Maize plants: an ideal production plat- combinant protein production. Biotechnol Lett 2010;32:137383.
form for effective and safe molecular pharming. Plant Sci 2008b;174:40919. Spencer D, Dickey LF, Gasdaska JR, Wang X, Cox KM, Peele CG. 2010. Expression of plas-
Ramessar K, Rademacher T, Sack M, Stadlmann J, Platis D, Stiegler G, et al. Cost-effective minogen and microplasminogen in duck weed. United States Patent, No. 7659445
production of a vaginal protein microbicide to prevent HIV transmission. Proc Natl Staub JM, Garcia B, Graves J, Hajdukiewicz PTJ, Hunter P, Nehra N, et al. High-yield pro-
Acad Sci U S A 2008c;105:372732. duction of a human therapeutic protein in tobacco chloroplasts. Nat Biotechnol
Ramessar K, Capell T, Twyman RM, Quemada H, Christou P. Calling the tunes on trans- 2000;18:3338.
genic crops: the case for regulatory harmony. Mol Breed 2009;23:99-112. Sticklen MB. 2007. Production of ss-glucosidase, hemicellulase and ligninase in E1 and
Rasala BA, Muto M, Lee PA, Jager M, Cardoso RMF, Behnke CA, et al. Production of ther- FLC-cellulase-transgenic plants. United States Patent Application, No. 20070192900
apeutic proteins in algae, analysis of expression of seven human proteins in the Stomp AM. The duckweeds: a valuable plant for biomanufacturing. Biotechnol Annu
chloroplast of Chlamydomonas reinhardtii. Plant Biotechnol J 2010;8:71933. Rev 2005;11:6999.
Rival S, Wisniewski JP, Langlais A, Kaplan H, Freyssinet G, Vancanneyt G, et al. Spirodela Strasser R, Stadlmann J, Schahs M, Stiegler G, Quendler H, Mach L, et al. Generation of
(duckweed) as an alternative production system for pharmaceuticals: a case study, glyco-engineered Nicotiana benthamiana for the production of monoclonal anti-
aprotinin. Transgenic Res 2008;17:50313. bodies with a homogeneous human-like N-glycan structure. Plant Biotechnol J
Rosenberg JN, Oyler GA, Wilkinson L, Betenbaugh MJ. A green light for engineered 2008;6:392402.
algae: redirecting metabolism to fuel a biotechnology revolution. Curr Opin Bio- Streateld SJ. Engineered chloroplasts as vaccine factories to combat bioterrorism.
technol 2008;19:4306. Trends Biotechnol 2006;24:33942.
Ruggiero F, Exposito JY, Bournat P, Gruber V, Perret S, Comte J, et al. Triple helix assem- Su WW, Lee KT. Plant cell and hairy-root cultures-process characteristics, products, and
bly and processing of human collagen produced in transgenic tobacco plants. FEBS application. In: Yang ST, editor. Bioprocessing for value-added products from renew-
Lett 2000;469:1326. able resources. Amsterdam, The Netherlands: Elsevier Science; 2007. p. 26392.
Ruhlman T, Verma D, Samson N, Daniell H. The role of heterologous chloroplast se- Surzycki R, Greenham K, Kitayama K, Dibal F, Wagner R, Rochaix JD, et al. Factors effect-
quence elements in transgene integration and expression. Plant Physiol ing expression of vaccines in microalgae. Biologicals 2009;37:1338.
2010;152:2088104. Tiwari S, Verma PC, Singh PK, Tuli R. Plants as bioreactors for the production of vaccine
Rukavtsova EB, Abramikhina TV, Shulga NY, Bykov VA. Bur'yanov YI. Tissue specic ex- antigens. Biotechnol Adv 2009;27:44967.
pression of hepatitis B virus surface antigen in Transgenic plant cells and tissue cul- Tran M, Zhou B, Pettersson PL, Gonzalez MJ, Mayeld SP. Synthesis and assembly of a
ture. Russ. J Plant Physiol 2007;54:7705. full-length human monoclonal antibody in algal chloroplasts. Biotechnol Bioeng
Rybicki EP. Plant-made vaccines for humans and animals. Plant Biotechnol J 2010;8: 2009;104:66373.
62037. Tremblay R, Wang D, Jevnikar AM, Ma S. Tobacco, a highly efcient green bioreactor for
Santi L, Giritch A, Roy CJ, Marillonnet S, Klimyuk V, Gleba Y, et al. Protection conferred production of therapeutic proteins. Biotechnol Adv 2010;28:21421.
by recombinant Yersinia pestis antigens produced by a rapid and highly scalable Tremblay R, Feng M, Menassa R, Huner NP, Jevnikar AM, Ma S. High-yield expression of
plant expression system. Proc Natl Acad Sci U S A 2006;103:8616. recombinant soybean agglutinin in plants using transient and stable systems.
Santi L, Batchelor L, Huang Z, Hjelm B, Kilbourne J, Arntzen CJ, et al. An efcient plant Transgenic Res 2011;20:34556.
viral expression system generating orally immunogenic Norwalk virus-like parti- Trexler MM, McDonald KA, Jackman AP. A cyclical semicontinuous process for produc-
cles. Vaccine 2008;26:184654. tion of human alpha(1)-antitrypsin using metabolically induced plant cell suspen-
Schaaf A, Tintelnot S, Baur A, Reski R, Gorr G, Decker EL. Use of endogenous signal se- sion cultures. Biotechnol Prog 2005;21:3218.
quences for transient production and efcient secretion by moss (Physcomitrella Twyman RM, Stoger E, Schillberg S, Christou P, Fischer R. Molecular farming in plants:
patens) cells. BMC Biotechnol 2005;5:30. host systems and expression technology. Trends Biotechnol 2003;21:5708.
Schahs M, Strasser R, Stadlmann J, Kunert R, Rademacher T, Steinkellner H. Production Vezina LP, Faye L, Lerouge P, D'Aoust MA, Marquet-Blouin E, Burel C, et al. Transient
of a monoclonal antibody in plants with a humanized N-glycosylation pattern. co-expression for fast and high-yield production of antibodies with human-like
Plant Biotechnol J 2007;5:65763. N-glycans in plants. Plant Biotechnol J 2009;7:44255.
Scheller J, Conrad U. Plant-based material, protein and biodegradable plastic. Curr Opin Voinnet O, Rivas S, Mestre P, Baulcombe D. An enhanced transient expression system in
Plant Biol 2005;8:18896. plants based on suppression of gene silencing by the p19 protein of tomato bushy
Scheller J, Guhrs KH, Grosse F, Conrad U. Production of spider silk proteins in tobacco stunt virus. Plant J 2003;33:94956.
and potato. Nat Biotechnol 2001;19:5737. Vunsh R, Li JH, Hanania U, Edelman M, Flaishman M, Perl A, et al. High expression of
Shaaltiel Y, Bartfeld D, Hashmueli S, Baum G, Brill-Almon E, Galili G, et al. Production of transgene protein in Spirodela. Plant Cell Rep 2007;26:15119.
glucocerebrosidase with terminal mannose glycans for enzyme replacement ther- Walker TL, Purton S, Becker DK, Collet C. Microalgae as bioreactors. Plant Cell Rep
apy of Gaucher's disease using a plant cell system. Plant Biotechnol J 2007;5: 2005;24:62941.
57990. Waltz E. Amylase corn sparks worries. Nat Biotechnol 2011;29:294.
Shanks JV, Morgan J. Plant hairy root culture. Curr Opin Biotechnol 1999;10:1515. Wang DJ, Brandsma M, Yin ZQ, Wang AM, Jevnikar AM, Ma SW. A novel platform for
Sharma AK, Sharma MK. Plants as bioreactors: recent developments and emerging op- biologically active recombinant human interleukin-13 production. Plant Biotech-
portunities. Biotechnol Adv 2009;27:81132. nol J 2008;6:50415.
Sharp JM, Doran PM. Strategies for enhancing monoclonal antibody accumulation in Wang L, Coppel RL. Oral vaccine delivery: can it protect against non-mucosal patho-
plant cell and organ cultures. Biotechnol Prog 2001;17:97992. gens? Expert Rev Vaccines 2008;7:72938.
Sheludko YV. Agrobacterium-mediated transient expression as an approach to produc- Weathers PJ, Giles KL. Regeneration of plants using nutrient mist culture. In Vitro Cell
tion of recombinant proteins in plants. Recent Pat Biotechnol 2008;2:198208. Dev Biol-Plant 1988;24:72432.
Sheludko YV, Sindarovska YR, Gerasymenko IM, Bannikova MA, Kuchuk NV. Compari- Weathers PJ, Towler MJ, Xu JF. Bench to batch: advances in plant cell culture for pro-
son of several Nicotiana species as hosts for high-scale Agrobacterium-mediated ducing useful products. Appl Microbiol Biotechnol 2010;85:133951.
transient expression. Biotechnol Bioeng 2007;96:60814. Weathers PJ, Towler MJ, Wyslouzil BE. Beyond cells: culturing complex plant tissues.
Shih SMH, Doran PM. Foreign protein production using plant cell and organ cultures: In: El-Mansi EMT, Bryce CFA, Demain AL, Allman AR, editors. Fermentation Micro-
advantages and limitations. Biotechnol Adv 2009;27:103642. biology and Biotechnology. Boca Raton, FL: CRC Press 3rd edition; in press.
Shin YJ, Hong SY, Kwon TH, Jang YS, Yang MS. High level of expression of recombinant Whaley KJ, Hiatt A, Zeitlin L, Hum Vaccin. Emerging antibody products and Nicotiana
human granulocytemacrophage colony stimulating factor in transgenic rice cell manufacturing. Hum Vaccin 2011:7.
suspension culture. Biotechnol Bioeng 2003;82:77883. Wilson JA, Hevey M, Bakken R, Guest S, Bray M, Schmaljohn AL, et al. Epitopes involved
Shin YJ, Lee NJ, Kim J, An XH, Yang MS, Kwon TH. High-level production of bioactive in antibody-mediated protection from Ebola virus. Science 2000;287:16646.
heterodimeric protein human interleukin-12 in rice. Enzyme MicrobTechnol Witcher DR, Hood EE, Peterson D, Bailey M, Bond D, Kusnadi A, et al. Commercial pro-
2010;46:34751. duction of -glucuronidase (GUS): a model system for the production of proteins
Shoji Y, Chichester JA, Bi H, Musiychuk K, de la Rosa P, Goldschmidt L, et al. Plant-ex- in plants. Mol Breed 1998;4:30112.
pressed HA as a seasonal inuenza vaccine candidate. Vaccine 2008;26:29304. Wongsamuth R, Doran PM. Production of monoclonal antibodies by tobacco hairy
Shoji Y, Bi H, Musiychuk K, Rhee A, Horsey A, Roy G, et al. Plant-derived hemagglutinin roots. Biotechnol Bioeng 1997;54:40115.
protects ferrets against challenge infection with the A/Indonesia/05/05 strain of Woodard SL, Mayor JM, Bailey MR, Barker DK, Love RT, Lane JR, et al. Maize (Zea
avian inuenza. Vaccine 2009;27:108792. mays)-derived bovine trypsin: characterization of the rst large-scale, commercial
Shoji Y, Chichester JA, Jones M, Manceva SD, Damon E, Mett V, et al. Plant-based rapid protein product from transgenic plants. Biotechnol Appl Biochem 2003;38:12330.
production of recombinant subunit hemagglutinin vaccines targeting H1N1 and Woods RR, Geyer BC, Mor TS. Hairy-root organ cultures for the production of human
H5N1 inuenza. Hum Vaccin 2011;7:4150. acetylcholinesterase. BMC Biotechnol 2008;8:95.
1184 J. Xu et al. / Biotechnology Advances 30 (2012) 11711184

Xie T, Qiu Q, Zhang W, Ning T, Yang W, Zheng C, et al. A biologically active rhIGF-1 fusion Yamamoto YT, Rajbhandari N, Lin XH, Bergmann BA, Nishimura Y, Stomp AM. Genetic
accumulated in transgenic rice seeds can reduce blood glucose in diabetic mice via transformation of duckweed Lemna gibba and Lemna minor. In Vitro Cell Dev Biol
oral delivery. Peptides 2008;29:186270. Plant 2001;37:34953.
Xu JF, Shpak E, Gu TY, Moo-Young M, Kieliszewski M. Production of recombinant plant Yang JJ, Barr LA, Fahnestock SR, Liu ZB. High yield recombinant silk-like protein pro-
gum with tobacco cell culture in bioreactor and gum characterization. Biotechnol duction in transgenic plants through protein targeting. Transgenic Res 2005;14:
Bioeng 2005;90:57888. 31324.
Xu JF, Tan L, Goodrum KJ, Kieliszewski MJ. High-yields and extended serum half-life of Yang L, Tada Y, Yamamoto MP, Zhao H, Yoshikawa M, Takaiwa F. A transgenic rice seed
human interferon alpha 2b expressed in tobacco cells as Arabinogalactan-protein accumulating an anti-hypertensive peptide reduces the blood pressure of sponta-
fusions. Biotechnol Bioeng 2007;97:997-1008. neously hypertensive rats. FEBS Lett 2006;580:331520.
Xu JF, Okada S, Tan L, Goodrum KJ, Kopchick JJ, Kieliszewski MJ. Human growth hor- Yang L, Wakasa Y, Takaiwa F. Biopharming to increase bioactive peptides in rice seed.
mone expressed in tobacco cells as an arabinogalactan-protein fusion glycoprotein J AOAC Int 2008;91:95764.
has a prolonged serum life. Transgenic Res 2010;19(5):84967. Yoon SM, Kim SY, Li KF, Yoon BH, Choe S, Kuo MM. Transgenic microalgae expres-
Xu JF, Ge XM, Dolan MC. Towards high-yield production of pharmaceutical proteins sing Escherichia coli AppA phytase as feed additive to reduce phytate excretion
with plant cell suspension cultures. Biotechnol Adv 2011a;29:27899. in the manure of young broiler chicks. Appl Microbiol Biotechnol 2011;91:
Xu L, Weathers PJ, Xiong XR, Liu CZ. Microalgal bioreactors: challenges and opportuni- 55363.
ties. Eng Life Sci 2009;9:17889. Yusibov VM, Mamedov TG. Plants as an alternative system for expression of vaccine
Xu X, Gan Q, Clough RC, Pappu KM, Howard JA, Baez JA, et al. Hydroxylation of recom- antigens. Proc ANAS (Biological Sciences) 2010;65:195200.
binant human collagen type I alpha 1 in transgenic maize co-expressed with a re- Yusibov V, Streateld SJ, Kushnir N. Clinical development of plant-produced recombi-
combinant human prolyl 4-hydroxylase. BMC Biotechnol 2011b;11:69. nant pharmaceuticals: vaccines, antibodies, and beyond. Hum Vaccin 2011;7:
Xue GP, Patel M, Johnson JS, Smyth DJ, Vickers CE. Selectable marker-free transgenic 31321.
barley producing a high level of cellulase (1,4-beta-glucanase) in developing Zhang Y, Hu Y, Yang B, Ma F, Lu P, Li L, et al. Duckweed (Lemna minor) as a model plant
grains. Plant Cell Rep 2003;21:108894. system for the study of human microbial pathogenesis. PLoS One 2010;5:e13527.

You might also like