Você está na página 1de 17

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 577

Plants as biofactories: Production of


pharmaceutical recombinant proteins
CHIARA LICO, ANGIOLA DESIDERIO, SELENE BANCHIERI AND EUGENIO BENVENUTO*
ENEA, UTS BIOTEC, Section of Genetics and Plant Genomics
C.R. Casaccia, Via Anguillarese, 301, 00060 S. Maria di Galeria, Rome, Italy
*Corresponding author: benvenutoe@casaccia.enea.it

Abstract
Plants can be used for the large-scale production of a variety of recombinant proteins, destined for agroindustrial, biomedical and pharmaceutical applications. In
fact, blood components, hormones, enzymes, cytokines, antibodies and vaccines
have been successfully produced in plants. Plant systems offer unique advantages in
comparison with conventional techniques (bacteria, mammal or yeast cells): i) lower
production costs; ii) synthesis of functional proteins, similar to those produced in
animal cells, absolutely free of animal pathogens; iii) easy scale-up and purification
technology. Furthermore, the engineering of edible plants may allow for the delivery
of the recombinant protein (e.g. vaccines) through fruits, tubers, leaves or seeds. In
this way the cold chain, necessary for the storage and the transport of purified
recombinant products could be avoided, as well as the administration procedures by
injection. This review describes gene transfer methods (including stable and transient transformation), plant species used and strategies to obtain high yields of protein, with attention focused towards plant-derived antibodies and vaccines, known
as plantibodies or plantigens. Up to now, several groups are working in this
promising field of research demonstrating that plants are able to produce proteins
derived from different kingdoms, with highly complex structures (e.g. immunoglobulins A and G) and the different, post-translational, modification patterns of plants
do not dramatically affect the properties or the biological activity of the recombinant
protein. Using plants as biofactories to produce green therapeutic proteins is not
only a proof of concept but a reality as an approach for agroindustry.

Introduction: Plants as biofactories


Genomic and proteomic approaches to the study of fundamental cell mechanisms
are rapidly contributing to broaden our knowledge of metabolic pathways for the
Tuberosa R., Phillips R.L., Gale M. (eds.), Proceedings of the International Congress
In the Wake of the Double Helix: From the Green Revolution to the Gene Revolution,
27-31 May 2003, Bologna, Italy, 577-593, 2005 Avenue media, Bologna, Italy.

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 578

LICO ET AL.

exploitation of the cell factory. In the last few years, this knowledge has led to
important advances in the large-scale production of diagnostic and therapeutic proteins in heterologous hosts (bacteria, yeasts, mammalian or insect cells, and transgenic animals and plants), allowing the comparison of the most efficient method in
terms of costs, product quality and safety.
The new branch of plant biotechnology defined as molecular farming is mainly focused on the exploitation of plants of agronomic relevance as factories for large
scale production of biomolecules. In particular, the possibility of producing biopharmaceuticals using plants, paves new ways and offers solutions to some of the
problems associated to traditional foreign-expression systems. For example, the production of complex multimeric proteins, such as antibodies, in bacteria is limited
by the absence of the complex enzymatic machinery involved in post-translational
modification of neo-synthesized proteins. Among eukaryotic expression systems,
yeast is not always appropriate as hyperglycosylation of the final product is often
encountered (Harashima 1994; Malissard et al. 1996). Insect and mammalian cell
cultures represent complex systems requiring cumbersome, expensive procedures
and may be easily contaminated with toxins, viruses or prions raising concerns
about the safety of the final product. Finally, transgenic animals pose analogous
risks, in addition to slow scaling-up (in terms of time required for reproduction)
and ethical concerns. Plant systems may represent the solution to most of these
problems. First of all, plant cells are able to produce proteins structurally and functionally equivalent to those synthesized in mammalian cells, guaranteeing
high-quality products free of human pathogens. A distinct advantage is represented by the possibility of using plants as natural-storing systems when the synthesis
of the biomolecule is properly targeted to seeds or storage organs, allowing its conservation at room temperature. Furthermore, it might be possible to directly deliver the product by oral administration of tissues (fruits, leaves and roots) of an edible plant. In this way, extraction and purification of the recombinant product is
avoided and the biomolecule is also protected by a sort of capsule (the cell wall) for
prolonged action on mucosal surfaces.
Finally, the established agronomic procedures of crops offer big economic
advantages, with the only drawbacks being represented by the control of production steps and containment of genetically modified plants. The first recombinant
protein to be synthesized in planta was the human growth hormone (Barta et al.
1986). Since then, many other proteins have been produced by plant systems
(Table 1) and some of them have been commercialized (Hood et al. 1997; Witcher et al. 1998) proving their efficacy and competitiveness compared to other systems (Evangelista et al. 1998; Kusnadi et al. 1998).
It is important to highlight that plants can also be used as biofactories of endogenous compounds (e.g. acetil-salicilic acid and opiates), by modifying the metabolic pathways to improve the natural expression levels. Seventy percent of the cur578

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 579

LICO ET AL.

Table 1. Recombinant proteins expressed in plants for pharmaceutical applications.


Year

Protein

Transgenic species

References

1986
1990
1990
1993

Human growth hormone


Human albumin
Human albumin
Epidermal growth factor

N. tabacum
N. tabacum
S. tuberosum
N. tabacum

Barta et al. 1986


Sijmons et al. 1990
Sijmons et al. 1990
Higo et al. 1993

1994

Human interferon-

O. sativa

Zhu et al. 1994

1995

Eritropoietin

N. tabacum

Matsumoto et al. 1995

1997

-casein

S. tuberosum

Chong et al. 1997

1997

and hemoglobin

N. tabacum

Dieryck et al. 1997

1997

Human mucarinic cholinergic receptors

N. tabacum

Mu et al. 1997

1998

Interleukine 2 and 4

N. tabacum

Magnuson et al. 1998

1999

Human 1-antitrypsin

O. sativa

Terashima et al. 1999

2000

Somatotropine

N. tabacum

Leite et al. 2000

2000

Collagen

N. tabacum

Ruggiero et al. 2000

2000

Lactoferrin

S. tuberosum

Chong et al. 2000

2000

Somatotropin

N. tabacum

Staub et al. 2000

2001

Human acetylcholinesterase

L. esculentum

Mor et al. 2001

2002

Bovin aprotinin

Z. mays

Azzoni et al. 2002

2002

Human collagen

N. tabacum

Merle et al. 2002

2003

Lactoferrin

N. tabacum

Choi et al. 2003

2003

Interleukine 18

N. tabacum

Zhang et al. 2003

2003

Human granulocyte-macrophage colony


stimulating factor

O. sativa

Shin et al. 2003

rently available pharmaceutical products are natural or naturally-derived compounds of plant origin. Molecular farming could potentially extend the range of
plant applications in the pharmaceutical industry, offering a valuable alternative to
other biological systems used to produce complex macromolecules.

Transformation methods
The expression of heterologous proteins in plants can be performed using two different methodologies. One is based on the stable expression of the recombinant
protein in transgenic plants and the other on the transient expression obtained
through epichromosomal transformation.
579

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 580

LICO ET AL.

Stable expression in nuclear or chloroplast genome

The most employed method is based on the natural capability of Agrobacterium


tumefaciens to stably transfer DNA into plant cells (Tinland 1996). The transformation of plant tissues by this technique can be performed either in vitro, on small
leaf explants, or directly in planta (Feldman and Marks 1987; Bechtold et al. 1993;
Clough and Bent 1998).
For plant species that are recalcitrant to Agrobacterium infection (i.e. monocots),
nuclear transformation can be achieved using the so-called biolistic method. This
technique consists of shooting tungsten or gold microbeads coated in DNA vector
through a particle gun on plant tissues. The biolistic method is also used for plastid transformation by inserting foreign DNA into the genome of chloroplasts. The
fundamental difference between plastid and nuclear transformation is that due to
homologous recombination in the chloroplast, the insertion of the foreign gene is in
a defined position in the organelles genome, often allowing us to obtain high protein
yields. The prokaryotic origin of the chloroplast poses some limitations, as complex
proteins require post-translational modifications that are not produced. Nonetheless,
this strategy is currently under rapid diffusion for biosafety reasons, as this organelle
is maternally inherited which reduces the risk of transgenes spreading through pollen.
Since the first attempts at gene transfer more than 20 years ago, the choice of
plant species was based on its genetic and physiological features such as in vitro
plasticity and its ease of transformation. Nowadays, several plant species can be
successfully transformed and research interests in molecular farming are directing the choice of several crops, food or non-food, depending on the final goal of
the project.

Factors influencing exogenous gene expression

One of the major prerequisites for the construction of plant biofactories with
good exogenous gene expression levels is the preparation of transformation vectors.
Among the factors to be considered are the codon usage and the flanking regulatory regions of the heterologous sequence. Usually, to reach high level of expression
constitutive promoters such as the CaMV (Cauliflower Mosaic Virus) 35S promoter (ODell et al. 1985) for most plants or the maize ubiquitin-1 promoter
(Christensen and Quail 1996) for monocots are used. The expression in specific
plant tissues/organs such as in seeds and in tubers or the expression in response to
specific physical or chemical inputs is possible by using tissue-specific or inducible
promoters (Zuo and Chua 2000). Also, viral translational enhancers can be useful
tools to increase the yield of the heterologous protein (Gallie and Walbot 1992). In
cereals, to increase the synthesis of the heterologous protein, an intron derived from
the maize polyubiquitin gene has been inserted in the coding region of the transgene (Vain et al. 1996). An important role is also attributed to polyadenilation signals generally derived from the 35S transcript of CaMV, the nopaline synthase gene
580

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 581

LICO ET AL.

(nos) of A. tumefaciens or the small subunit of the ribulose-1,5-biphosphate carboxilase (ssu) of Pisum.
Since stable nuclear transformation of plants implies the random insertion of the
transgene in the genome, the level of expression may be influenced by the transcriptional activity of the surrounding chromosomic region. Furthermore, the insertion
can hinder the transcription of genes involved in fundamental morpho-physiological
pathways producing atypical or lethal phenotypes. For these reasons, molecular characterization of transformants (e.g. evaluation of the copy number of the transgene and
definition of its position within the plant nuclear genome) is fundamental. The insertion of a high number of copies is not always an advantage, as high levels of expression can induce the activation of mechanisms of gene silencing, resulting in the total
suppression of the transgene (Metzlaff 2002). Plastid transformation could represent
a solution in that it eliminates positional effects and any possible interference on genes
involved in vital cell functions. Furthermore, activation of gene silencing has not been
described in transplastomic plants, despite the high yield of exogenous protein due to
the high number of transformed chloroplasts per single cell. Unfortunately, not all
plant species can be transformed with this technique (up to now only tobacco and
tomato) and once the transformation have been done it is very difficult to reach the
homoplasmic condition.
The quality of the product can be improved by targeting the protein to the secretory pathway of the cell where it can fold correctly and post-translational modifications can be introduced. Retention to the cell compartments can be obtained by
adding defined signal peptides, such as the H/KDEL peptide (Conrad and Fiedler
1998). This sequence is used to retain the proteins in the endoplasmic reticulum, such
as in the case where it is detrimental to proceed further in the pathway (i.e. the Golgi
apparatus), to avoid unwanted plant-specific glycosilation (Gomord and Faye 2004).
In fact, sugar residues added to proteins in the secretory pathway of plant cells are different from those of mammalian cells (presence of xylose and fucose and absence of
galactose and syalic acid). This aspect could have functional and immunologic implications (e.g. induction of allergic response; Bardor et al. 2003), even if recent data
demonstrate the absence of side effects on humans, induced by new glycosilation pattern of the foreign protein (Chargelegue et al. 2000).
Targeting of proteins to the secretory pathway can be also used to direct their
secretion through roots (rhyzosecretion). This strategy, requiring the growth of
plants in hydroponic cultures, can appreciably simplify the purification steps
(Drake et al. 2003).

Epichromosomal expression

The epichromosomal expression does not imply the integration of heterologous


sequences in the plant genome (transient transformation) and, as a consequence,
does not allow transgene inheritance, thus restricting the synthesis of the foreign
581

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 582

LICO ET AL.

molecule to a single plant. The most popular and widely-used technique for transient transformation is based on the use of genetically-modified plant viruses as vectors of the heterologous sequence. Transient transformation is an easy and rapid
strategy of expression, ensuring very high yields of the product of interest. This is
due to the fast multiplication of the modified virus in plant cells resulting in accumulation of exogenous sequence in the complex of viral expression machinery.
Usually, for single strand positive RNA viruses, such as Potato Virus X, the heterologous sequence is inserted into the viral genome as an additional open reading
frame (ORF) whose translation is controlled by a duplication of viral subgenomic
promoter (Figure 1b). Since the duplication of subgenomic promoters can sometimes result in recombination events leading to the loss of the exogenous sequence
(Chapman et al. 1992), alternative cloning strategies have been developed. For
instance, attaching the exogenous gene to an IRES sequence (Internal Ribosome
Entry Sequence; Toth et al. 2001) allowing the independent synthesis of two proteins coded by a bicistronic mRNA. This new fragment can then be inserted

Figure 1. Strategies used to obtain the transient expression of a sequence of interest (black box)
through viral vectors.

582

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 583

LICO ET AL.

between the subgenomic promoter for the coat protein gene (CP) and the CP gene
itself (Figure 1c) to be successfully expressed.
A possible limit to transient expression strategy could be the restricted host range
of the virus. Nonetheless, many plant viruses have been used for epichromosomal
transformation: Bromovirus, Hordivirus (BSMV: Barley Stripe Mosaic Virus),
Tobamovirus (TMV: Tobacco Mosaic Virus), Potyvirus, Potexvirus (PVX: Potato
Virus X), Comovirus (CPMV: Cowpea Mosaic Virus), Tombusvirus (TBSV: Tomato Bushy Stunt Virus) and Alfamovirus (AMV: Alfalfa Mosaic Virus). These viruses infect a wide range of plant species (Pogue et al. 2002).
An alternative strategy to virus-induced transient expression is the so-called
Agroinfiltration. This technique is typically used to evaluate the quality of an
expression construct in terms of yield, conformation and functionality of the
desired product, before the production of stably transformed plants. Moreover, it
offers the unique opportunity to simultaneously express the genes encoding for the
subunits of multimeric proteins, providing the possibility to evaluate their assembly and functionality (Vaquero et al. 1999). Agroinfiltration is based on the use of
A. tumefaciens, like stable transformation, but does not necessarily imply the integration of the exogenous DNA into the plant genome.

Plants expressing antibodies


Antibodies are key molecules of the vertebrate immune system. The typical structure
of these glycoproteins consists of four polypeptides, two heavy chains and two light
chains, linked together by disulfide bridges and non-covalent bonds. These complex
proteins are involved in the specific recognition of antigens (i.e. substances extraneous to the organism able to induce the activation of the immune response) by recognising a molecular structure with high-binding affinity and specificity. For these features, antibodies can be considered ideal reagents useful for biomedical and industrial applications, both in therapy and in molecular diagnostics. In fact, antibodies are
successfully used in clinical trials to direct drugs, radioisotopes and interfering
molecules to unhealthy cells and tissues (e.g. towards a tumour) and to obtain targeted therapeutic effects, reducing alteration of other organs. Moreover, the possibility
to deliver antibody inside cells to immunomodulate pathology-related functions
has been demonstrated. Finally, in diagnostics approaches, properly targeted antibodies allow for the identification, characterization and localization of physiological or
pathological alterations (Peeters et al. 2001; Ross et al. 2003).
The possibility of obtaining antibody expression in plants (plantibodies) was
first demonstrated by Hiatt et al. in 1989. In that case, the assembling and the correct folding of a full-length immunoglobulin type G in a non-animal cellular system required critical experimental approaches. Since then, different forms of antibodies have been successfully expressed in planta.
583

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 584

LICO ET AL.

Although the production of complete antibody molecules is essential for therapeutic applications requiring the activation of the immune system, many other
uses require only the preservation of the antigen binding site. The domain structure of immunoglobulins allows for the design and construction by molecular
engineering of different forms of recombinant antibodies resulting in the production of smaller and versatile molecules (Nolke et al. 2003; Kipriyanov and Le
Gall 2004). Several of these molecules, such as single domain antibodies (dAb;
Benvenuto et al. 1991), Fab fragments (De Neve et al. 1993) and single chain
variable fragments (scFv; De Wilde et al. 1999; Fiedler et al. 1999) have been successfully expressed in plants. The scFv fragments consist of immunoglobulin variable domains, connected by a linker peptide. These recombinant molecules
showed a great applicability due both to their easy expression in heterologous systems and versatility as reagents. Plants also represent the ideal production system
for fully multivalent antibodies, such as the secretory IgA (SigA; Ma et al. 1995).
SIgA molecules result from the expression and the assembly, through transgenic
plant crossing, of four polypeptide chains to form a dimeric IgA antibody, which
is particularly resistant to proteolysis. The first example of SIgA expressed in
plants has been the GUYS 13 antibody specific for the adhesion protein of Streptococcus mutans (the causative agent of dental caries). GUYS 13 has been successfully used in clinical trials, demonstrating the validity of plant-derived antibody for passive immunotherapy (Ma et al. 1998).
The antibody expression levels in transgenic plant system are variable (up to 5%
of total protein), depending on the plant species used and on the tissue where it is
expressed (Fiedler et al. 1997). Expression levels can be further improved using
transient plant transformation mediated by viral vectors (Fisher et al. 1999).
The expression of full-length immunoglobulins and antibody fragments can be
targeted to different plant cell compartments such as the apoplast, endoplasmic
reticulum and cytoplasm (Conrad and Fiedler 1998). This possibility may represent
an advantage for purification. Also, the ability of antibodies to specifically bind to
antigens can be useful for purification through chromatography procedures. The
purity level of plantibodies depends on their intended use. When antibodies are
designed for intravascular immunotherapy or for fine diagnostics, an accurate
purification is required. In contrast, in the case of topical passive immunization, the
use of edible plant-expressed antibodies is conceivable, reducing or completely
eliminating the purification procedure (De Wilde et al. 1999).
Many full-length or recombinant plantibodies produced either by academic
groups or companies have been used as therapeutic molecules. Some examples are
listed in Table 2. In the future, it is plausible that the use of plants to produce a high
amount of safe and highly-specific reagents, such as antibodies, will be further
improved.

584

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 585

LICO ET AL.

Table 2. Examples of pharmaceutical antibodies expressed in plants.


Antibody

Antigen specificities

Transgenic
species

Application

References

IgG and
SIgA

Streptococcus mutans

N. tabacum

Therapy (topical)

Ma et al. 1998

IgG

Colon cancer antigen

N. tabacum

Therapy/Diagnostics

Verch et al. 1998

IgG

Herpes Simplex
Virus (HSV-2)

G. max
O. sativa

Therapy (topical)

Zeitlin et al. 1998

Z. mays

Therapy (inhalation)

EPIcyte

Humanized Respiratory Syncytial


IgG
Virus
IgG

Clostridium difficile

Z. mays

Therapy (oral)

EPIcyte

IgG

Sperm

Z. mays

Contraceptive
(topical)

ReProtect LLC, MD

IgG

Hepatitis B antigen

N. tabacum

Therapy/Diagnostics

Valdes et al. 2003

Diabody

Carcinoembryonic
Antigen (CEA)

N. tabacum

Therapy/Diagnostics

Vaquero et al. 2002

scFv

Non-Hodgkins
(NHL) Lymphoma

N. tabacum

Vaccine

McCormick et al. 1999

scFv

Carcinoembryonic
Antigen (CEA)

N. tabacum, Therapy/Diagnostics
P. sativum,
L. esculentum,
O. sativa,
Triticum sp.

Stoger et al. 2000

Plants as systems for vaccine production and delivery


The formulation of subunit vaccines is based on pathogen-derived macromolecules
(proteins or peptides) known to induce protective immune responses without
inducing the disease. The production of these antigenic molecules in plants has all
the advantages of the expression of biopharmaceuticals in plant systems described
earlier, with the additional opportunity to get the expression in edible plant species
for oral administration of the vaccine without the need of purification treatments
(Walmsley and Arntzen 2003).
The production of vaccine components in plants can be achieved by both stable
and transient expression (Mason et al. 2002).
Stable expression has been exploited to express several antigens in different plant
species (e.g. potato, lettuce, lupin, tomato, carrot and maize; Table 3). Three of
585

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 586

LICO ET AL.

these antigen-expressing plants are at the moment under clinical trial to evaluate in
humans the efficacy of oral immunization by using transformed plant tissues. The
plants currently under study are potato, expressing the B subunit of E. coli
heat-labile toxin (Tacket et al. 1998) or the caspid protein of Norwalk virus (resulting in virus-like particles (VLPs) assembly in plant tissues; Tacket et al. 2000) and
lettuce expressing the major coat protein (HBsAg) of Hepatitis B virus (Kapusta et
al. 1999). Despite these promising results, until now it has not been possible to estimate the antigen delivered dose of immunization. This is mainly due to the variation of the expression levels of the heterologous gene among different transgenic
plants. Oral delivery of transgenic tissues is able to stimulate antibody titers similar
to those induced by traditional vaccines and present the additional advantage of
activating both systemic and mucosal immune responses.
Also, transient expression technology can be a useful strategy to synthesize vaccines
in plants. Aside from the production of antigenic proteins by inserting the complete
Table 3. Representative antigens expressed in transgenic plants.
Pathogen

Antigen

Transgenic specific

Human hepatitis B
virus

Coat protein epitope

N. tabacum
S. tuberosum
Lupinus luteus
Lactuca sativa

Mason et al. 1992


Thanavala et al. 1995
Richter et al. 2000
Ehsani et al. 1997
Kapusta et al. 1999

Norwalk virus

Coat Protein

N. tabacum
S. tuberosum

Mason et al. 1996


Tacket et al. 2000

Enterotoxic E. coli

Heat-labile enterotoxin B
subunit

N. tabacum
S. tuberosum

Haq et al. 1995


Mason et al. 1998
Tacket et al. 1998

Rabies virus

Glycoprotein

L. esculentum

McGarvey et al. 1995

Cytomegalovirus

Glycoprotein B

N. tabacum

Tackaberry et al. 1999

Vibrio cholerae

Cholera toxin B subunit

S. tuberosum

Arakawa et al. 1997, 1998

N. tabacum
S. tuberosum
D. carota

Ma et al. 1997
Avesani et al. 2003

Diabetes-associated
autoantigen

References

Foot-and-mouth
disease virus

Structural protein VP1

A. thaliana
M. sativa

Carrillo et al. 1998

Porcine transmissible
gastroenteritis
coronavirus

Glycoprotein S

A. thaliana
N. tabacum
Z. mays

Gomez et al. 1998


Tuboly et al. 2000
Streatfield et al. 2001

586

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 587

LICO ET AL.

coding sequences as additional ORFs in viral genomes (Figures 1b and 1c; Franconi
et al. 2002), purified chimeric viral particles can also be used as primary vaccine components (Table 4). By this approach, the sequence coding for an antigenic epitope (i.e.
the portion of an antigen, generally comprised of a few amino acids, responsible for
the activation of the immune response) is fused with the sequence encoding the coat
protein of a plant virus, in a position that is known to be displayed on the viral surface (Johnson et al. 1997; Figure 1d). In some instances, for example when using
Potato Virus X, long epitope sequences can also be expressed (more than 15-20 amino
acids) by inserting between the coat protein and the epitope sequences an additional
Table 4. Representative antigens expressed in plants through viral vector approach.
Pathogen

Antigen

Plant virus

References

Plasmodium
falciparum

Several epitopes

TMV

Turpen et al. 1995

Influenza virus

Hemagglutinin epitope

TMV

Sugiyama et al. 1995

Human
Immunodeficiency
Virus 1 (HIV-1)

Capsid epitopes

PVX
CPMV

Porta et al. 1996


Yusibov et al. 1997
Marusic et al. 2001

S. aureus

Fibronectin binding protein


(FNBP) epitope

PVX

Brennan et al. 1999

Hepatitis B virus

Mimotope

TMV

Nemchinov et al. 2000

Rabies virus

Chimeric peptide

AIMV

Yusibov et al. 2002

Human Papilloma
Virus (HPV)

E7 Oncoprotein

PVX

Franconi et al. 2002

Colerectal cancer

GA733-2 Antigen

TMV

Verch et al. 2004

Hepatitis C virus

Mimotope

CMV

Natilla et al. 2004

sequence coding for the 2A peptide of Foot and Mouth Disease Virus (FMDV; Ryan
et al. 1991; Ryan and Drew 1994). This peptide mediates an autocatalytic, stochastic
processing event that leads to the accumulation in the plant cell of a pool of both
recombinant and wildtype coat proteins (Santa Cruz et al. 1996; Figure 1e), and the
consequent virion displays both kinds of proteins.
Chimeric virus particles (CVPs) produced in plant tissues by using these strategies
have been able to induce in animal models strong antibody responses also without
adjuvants delivery (i.e. substances generally used in vaccine formulations to potentiate the immune response; Yusibov et al. 1997; Marusic et al. 2001). The high
immunogenicity of this kind of vaccines can be related to the high number of epitopes that are displayed on the surface of a complex molecular structure such as a virus
particle. Additional results from studies focused on the oral delivery in humans (phase
587

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 588

LICO ET AL.

I clinical trial) of spinach leaves infected with CVP displaying a rabies virus-derived
peptide (Yusibov et al. 2002) further strengthened the idea that transient expression
can also be considered as a useful tool for vaccine production in plants.

Conclusions
Plants are good expression systems, and in some aspects can be considered more
efficient biofactories than bacteria or yeast/mammalian cells for the production of
high value molecules. Plants have been used for the large-scale production of several recombinant proteins, especially for biopharmaceutical applications with particular emphasis on antibodies and vaccine components. Applications of these
plant-produced recombinant molecules imply that they must be considered similarly to the traditional pharmaceuticals before their introduction to the market. To
reach this goal it is fundamental to cultivate these plants in contained greenhouses
for a complete control of the productive row, before entering the standardized
Good Laboratory Practice (GLP) of the pharmaceutical industry.

References
Arakawa T, Chong DK, Merritt JL, Langridge WH (1997) Expression of cholera toxin B subunit
oligomers in transgenic potato plants. Transgenic Res 6:403-413
Arakawa T, Chong DK, Langridge WH (1998) Efficacy of a food plant-based oral cholera toxin B
subunit vaccine. Nat Biotechnol 16:292-297
Avesani L, Falorni A, Tornielli GB, Marusic C, Porceddu A, Polverari A, Faleri C, Calcinaro F, Pezzotti M (2003) Improved in planta expression of the human islet autoantigen glutamic acid
decarboxylase (GAD65). Transgenic Res 12:203-212
Azzoni AR, Kusnadi AR, Miranda EA, Nikolov ZL (2002) Recombinant aprotinin produced in
transgenic corn seed: extraction and purification studies. Biotechnol Bioeng 80:268-276
Bardor M, Faveeuw C, Fitchette AC, Gilbert D, Galas L, Trottein F, Faye L, Lerouge P (2003)
Immunoreactivity in mammals of two typical plant glyco-epitopes, core alpha(1,3)-fucose and
core xylose. Glycobiol 13:427-34
Barta A, Sommergruber K, Thompson D, Harmuth K, Matzke ME, Matzke AJM (1986) The
expression of nopaline synthase-human growth hormone chimeric gene in transformed tobacco
and sunflower callus tissue. Plant Mol Biol 6:347-357
Bechtold N, Ellis J, Pelletier G (1993) In planta Agrobacterium mediated gene transfer by infiltration of adult Arabidopsis thaliana plants. CR Acad Sci Paris, Life Sci 316:1194-1199
Benvenuto E, Ordas RJ, Tavazza R, Ancora G, Biocca S, Cattaneo A, Galeffi P (1991) Phytoantibodies: a general vector for the expression of immunoglobulin domains in transgenic plants.
Plant Mol Biol 17:865-874
Brennan FR, Gilleland LB, Staczek J, Bendig MM, Hamilton WD, Gilleland HEJr (1999) A chimaeric plant virus vaccine protects mice against a bacterial infection. Microbiol
145:2061-2067

588

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 589

LICO ET AL.

Carrillo C, Wigdorovitz A, Oliveros JC, Zamorano PI, Sadir AM, Gomez N, Salinas J, Escribano
JM, Borca MV (1998) Protective immune response to foot-and-mouth disease virus with VP1
expressed in transgenic plants. J Virol 72:1688-1690
Chapman S, Kavanagh T, Baulcombe D (1992) Potato Virus X as a vector for gene expression in
plants. Plant J 2:549-557
Chargelegue D, Vine ND, van Dolleweerd CJ, Drake PM, Ma JK (2000) A murine monoclonal
antibody produced in transgenic plants with plant-specific glycans is not immunogenic in mice.
Transgenic Res 9:187-194
Choi SM, Lee OS, Kwon SY, Kwak S.S., Yu DY, Lee HS (2003) High expression of a human lactoferrin in transgenic tobacco cell cultures. Biotechnol Lett 25:213-218
Chong DK, Roberts W, Arakawa T, Illes K, Bagi G, Slattery CW, Langridge WH (1997) Expression of the human milk protein beta-casein in transgenic potato plants. Transgenic Res
6:289-296
Chong DK, Langridge WH (2000) Expression of full-length bioactive antimicrobial human lactoferrin in potato plants. Transgenic Res 9:7178
Christensen AH, Quail PH (1996) Ubiquitin promoter-based vectors for high level expression of
selectable and/or screenable marker genes in monocotyledonous plants. Transgenic Res
5:213-218
Clough SJ, Bent AF (1998) Floral dip: a simplified method for Agrobacterium-mediated transformation of Arabidopsis thaliana. Plant J 16:735-743
Conrad U, Fiedler U (1998) Compartment-specific accumulation of recombinant immunoglobulins in plant cells: an essential tool for antibody production and immunomodulation of physiological functions and pathogen activity. Plant Mol Biol 38:101-109
De Neve M, De Loose M, Jacobs A, Van Houdt H, Kaluza B, Weidle U, Van Montagu M, Depicker A (1993) Assembly of an antibody and its derived antibody fragment in Nicotiana and Arabidopsis. Transgenic Res 2:227-237
De Wilde C, De Jaeger G, De Neve M, van Montagu M, Depicker A (1999) Production of antibodies in transgenic plants A general Introduction. In: Harper K, Ziegler A (eds) Recombinant antibodies: applications in plant pathology. Taylor and Francis, London, UK pp 113-127
Dieryck W, Pagnier J, Poyart C, Marden MC, Gruber V, Bournat P, Baudino S, Merot B (1997)
Human haemoglobin from transgenic tobacco. Nature 386:29-30
Drake PM, Chargelegue DM, Vine ND, van Dolleweerd CJ, Obregon P, Ma JK (2003) Rhizosecretion of a monoclonal antibody protein complex from transgenic tobacco roots. Plant Mol
Biol 52:233-241
Ehsani P, Khabiri A, Domansky NN (1997) Polypeptides of hepatitis B surface antigen produced
in transegnic potato. Gene 190:107-111
Evangelista RL, Kusnadi AR, Howard JA, Nikolov ZL (1998) Process and economic evaluation of
the extraction and purification of recombinant beta-glucuronidase from transgenic corn.
Biotechnol Prog 14:607-614
Feldman K, Marks M (1987) Agrobacterium-mediated transformation of germinating seeds od Arabidopsis thaliana: a non-tissue culture approach. Mol Gen Genet 208:1-9
Fiedler U, Phillips J, Artsaenko O, Conrad U (1997) Optimization of scFv antibody production in
transgenic plants. Immunotechnol 3:205-216
Fiedler U, Artsaenko O, Phillips J, Conrad U (1999) Transgenic plants A low cost production system
of recombinant antibodies. In: Harper K, Ziegler A (eds) Recombinant antibodies: applications in
plant pathology. Taylor and Francis, London, UK pp 129-144

589

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 590

LICO ET AL.

Fisher R, Vaquero-Martin C, Sack M, Drossard J, Emans N, Commandeur U (1999) Towards


molecular farming in the future: transient protein expression in plant. Biotechnol Appl Bioc
30:113-116
Franconi R, Di Bonito P, Dibello F, Accardi L, Muller A, Birilli A, Simeone P, Don MG, Venuti
A, Giorni C (2002) Plant-derived Human Papillomavirus 16 E7 oncoprotein induces immune
response and specific tumor protection. Cancer Res 62:3654-3658
Gallie DR, Walbot V (1992) Identification of the motifs within the tobacco mosaic virus 5-leader
responsible for enhancing translation. Nucleic Acid Res 17:4631-4638
Gomez N, Carrillo C, Salinas J, Parra F, Borca MV, Escribano JM (1998) Expression of immunogenic glycoprotein S polypeptides from transmissible gastroenteritis coronavirus in transgenic
plants. Virology 249:352-358
Gomord V, Faye L (2004) Posttranslational modification of therapeutic proteins in plants. Curr
Opin Plant Biol 7:171-181
Harashima S (1994) Heterologous protein production by yeast-host vector systems. Bioprocess
Technol 19:137-158
Haq TA, Mason HS, Clements JD, Arntzen CJ (1995) Oral immunization with a recombinant bacterial antigen produced in transgenic plants. Science 268:714-716
Hiatt AC, Cafferkey R, Bowdish K (1989) Production of antibodies in transgenic plants. Nature
342:76-78
Higo K, Saito Y, Higo H (1993) Expression of a chemically synthesized gene for human epidermal
growth factor under the control of cauliflower mosaic virus 35S promoter in transgenic tobacco. Biosci Biotech Bioch 57:1477-1481
Hood EE, Witcher DR, Maddock S, Meyer T, Baszczynski C, Bailey M, Flynn P, Register J, Marshall L, Bond D, Kulisek E, Kusnadi A, Evangelista R, Nikolov Z, Wooge C, Mehigh RJ, Hernan R, Kappel WK, Ritland D, Li CP, Howard JA (1997) Commercial production of avidin
from transgenic maize: characterization of transformant, production, processing, extraction and
purification. Mol Breed 3:291-306
Johnson J, Lin T, Lomonossoff G (1997) Presentation of heterologous peptides on plant viruses:
genetics, structure and function. Ann Rev Phytopathol 35:67-86
Kapusta J, Modelska A, Figlerowicz M, Pniewski T, Letellier M, Lisowa O, Yusibov V, Koprowski
H, Plucienniczak A, Legocki AB (1999) A plant-derived edible vaccine against hepatitis B virus.
FASEB J 13:1796-1799
Kipriyanov SM, Le Gall F (2004) Generation and production of engineered antibodies. Mol
Biotechnol 26:39-60
Kusnadi AR, Evangelista RL, Hood EE, Howard JA, Nikolov ZL (1998) Processing of transgenic corn
seed and its effect on the recovery of recombinant beta-glucuronidase. Biotechnol Bioeng 60:44-52
Leite A, Kemper E, da Silva M, Luchessi A, Siloto R, Bonaccorsi E, El-Dorry H, Arruda P (2000)
Expression of correctly processed human growth hormone in seeds of transgenic tobacco plants.
Mol Breeding 6:47-53
Ma JK, Hiatt A, Hein M, Vine ND, Wang F, Stabila P, van Dolleweerd C, Mostov K, Lehner T
(1995) Generation and assembly of secretory antibodies in plants. Science 268:716-719
Ma SW, Zhao DL, Yin ZQ, Mukherjee R, Singh B, Qin HY, Stiller CR, Jevnikar AM (1997) Transgenic plants expressing autoantigens fed to mice to induce oral immune tolerance. Nat Med
3:793-796
Ma JK, Hikmat BY, Wycoff K, Vine ND, Chargelegue D, Yu L, Hein MB, Lehner T, (1998)
Characterization of a recombinant plant monoclonal secretory antibody and preventive
immunotherapy in humans. Nat Med 4:601-606

590

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 591

LICO ET AL.

Magnuson N., Linzmaier PM, Reeves R, An G, HayGlass K, Lee JM (1998) Secretion of biologically active human interleukin-2 and interleukin-4 from genetically modified tobacco cells in
suspension cultures. Protein Expres Purif 13:45-52
Malissard M, Borsig L, Di Marco S, Grutter MG, Kragl U, Wandrey C, Berger EG (1996) Recombinant soluble beta-1,4-galactosyltransferases expressed in Saccharomyces cerevisiae. Purification,
characterization and comparison with human enzyme. Eur J Biochem 239:340-348
Marusic C, Rizza P, Lattanzi L, Mancini C, Spada M, Belardelli F, Benvenuto E, Capone I (2001)
Chimeric plant virus particle as immunogens for inducing murine and human immune response
against Human Immunodeficiency virus Type 1. J Virol 75:8434-8439
Mason HS, Lam DMK, Arntzen CJ (1992) Expression of hepatitis B surface antigen in transgenic
plants. Proc Natl Acad Sci USA 89:11745-11749
Mason HS, Ball JM, Shi JJ, Jiang X, Estes MK, Arntzen CJ (1996) Expression of Norwalk capsid
protein in transgenic tobacco and potato and its oral immunogenicity in mice. Proc Natl Acad
Sci USA 93:5335-5340
Mason HS, Haq TA, Clements JD, Arntzen CJ (1998) Edible vaccine protects mice against
Escherichia coli heat-labile enterotoxin (LT): potatoes expressing a synthetic LT-B gene. Vaccine
16:1336-1343
Mason HS, Warzecha H, Mor T, Arntzen J (2002) Edible plant vaccines: applications for prophylactic and therapeutic molecular medecine. Trends Mol Med 8:324-329
Matsumoto S, Ikura K, Ueda M, Sasaki R (1995) Characterization of a human glycoprotein (erythropoietin) produced in cultured tobacco cells. Plant Mol Biol 27:1163-1172
McGarvey PB, Hammond J, Dienelt MM, Hooper DC, Fu ZF, Dietzschold B, Koprowski H,
Michaels FH (1995) Expression of the rabies virus glycoprotein in transgenic tomatoes.
Bio/Technol 13:1484-1487
McCormick AA, Kumagai MH, Hanley K, Turpen TH, Hakim I, Grill LK, Tuse D, Levy S, Levy
R (1999) Rapid production of specific vaccines for lymphoma by expression of the
tumor-derived single-chain Fv epitopes in tobacco plants. Proc Natl Acad Sci USA
96:703-708
Merle C, Perret S, Lacour T, Jonval V, Hudaverdian S, Garrone R, Ruggiero F, Theisen M (2002)
Hydroxylated human homotrimeric collagen I in Agrobacterium tumefaciens-mediated transient
expression and in transgenic tobacco plants. FEBS Lett 515:114-118
Metzlaff M (2002) RNA-mediated RNA degradation in transgene- and virus-induced gene silencing. Biol Chem 383:1483-1489
Mor TS, Sternfeld M, Soreq H, Arntzen CJ, Mason HS (2001) Expression of recombinant human
acetylcholinesterase in transgenic tomato plants. Biotechnol Bioeng 75:259-266
Mu JH, Chua NH, Ross EM (1997) Expression of human muscarinic cholinergic receptors in
tobacco. Plant Mol Biol 34:357-362
Natilla A, Piazzolla G, Nuzzaci M, Saldarelli P, Tortorella C, Antonaci S, Piazzolla P (2004) Cucumber mosaic virus as carrier of a hepatitis C virus-derived epitope. Arch Virol 149:137-154
Nemchinov LG, Liang TJ, Rifaat MM, Mazyad HM, Hadidi A, Keith JM (2000) Development of
a plant-derived subunit vaccine candidate against hepatitis C virus. Arch Virol 145:2557-2573
Nolke G, Fischer R, Schillberg S (2003) Production of therapeutic antibodies in plants. Expert
Opin Biol Th 3:1153-1162
ODell JT, Nagy F, Chua N-H (1985) Identification of DNA sequences required for activity of the
cauliflower mosaic virus 35S promoter. Nature 313:810-812
Peeters K, De Wilde C, De Jaeger G, Angenon G, Depicker A (2001) Production of antibodies and
antibody fragments in plants. Vaccine 19:2756-2761

591

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 592

LICO ET AL.

Pogue GP, Lindbo JA, Garger SJ, Fitzmaurice WP (2002) Making an ally from an enemy: plant
virology and the new agriculture. Annu Rev Phytopathol 40:45-74
Porta C, Spall VE, Lin T, Johnson JE, Lomonosoff GP (1996) The development of cowpea mosaic
virus as a potential source of novel vaccines. Intervirology 39:79-84
Richter LJ, Thanavala Y, Arntzen CJ, Mason HS (2000) Production of hepatitis B surface antigen
in transgenic plants for oral immunization. Nat Biotechnol 18:1167-1171
Ross JS, Gray K, Gray GS, Worland PJ, Rolfe M (2003) Anticancer antibodies. Am J Clin Pathol
119:472-485
Ruggiero F, Exposito J-Y, Bournat P, Gruber V, Perret S, Comte J, Olagnier B, Garrone R, Theisen
M (2000) Triple helix assembly and processing of human collagen produced in transgenic tobacco plants. FEBS Lett 469:132-136
Ryan MD, King AM, Thomas GP (1991) Cleavage of foot-and-mouth disease virus polyprotein is
mediated by residues located within a 19 amino acid sequence. J Gen Virol 72:2727-2732
Ryan MD, Drew J (1994) Foot-and-mouth disease virus 2A oligopeptide mediated cleavage of an
artificial polyprotein. EMBO J 13:928-933
Santa Cruz S, Chapman S, Roberts AG, Roberts IM, Prior DAM, Oparka KJ (1996) Assembly and
movement of a plant virus carrying a green fluorescent protein overcoat. Proc Natl Acad Sci USA
93:6286-6290
Shin YJ, Hong SY, Kwon TH, Jang YS, Yang MS (2003) High level of expression of recombinant
human granulocyte-macrophage colony stimulating factor in transgenic rice cell suspension culture. Biotechnol Bioeng 82:778-783
Sijmons PC, Dekker BMM, Schrammeijer B, Verwoerd TC, van den Elzen PJM, Hoekema A
(1990) Production of correctly processed human serum albumin in transgenic plants. Bio/Technol 8:217-221
Staub JM, Garcia B, Graves J, Hajdukiewicz PT, Hunter P, Nehra N, Paradkar V, Schlittler M, Carroll JA, Spatola L, Ward D, Ye G, Russell DA (2000) High-yield production of a human therapeutic protein in tobacco chloroplasts. Nat Biotechnol 18:333-338
Stoger E, Vaquero C, Torres E, Sack M, Nicholson L, Drossard J, Williams S, Keen D, Perrin Y,
Christou P, Fischer R (2000) Cereal crops as viable production and storage systems for pharmaceutical scFv antibodies. Plant Mol Biol 42:583-590
Streatfield SJ, Jilka JM, Hood EE, Turner DD, Bailey MR, Mayor JM, Woodard SL, Beifuss KK,
Horn ME, Delaney DE, Tizard IR, Howard JA (2001) Plant-based vaccines: unique advantages.
Vaccine 19: 2742-2748
Sugiyama Y, Hamamoto H, Takemoto S, Watanabe Y, Okada Y (1995) Systemic production of foreign peptides on the particle surface of tobacco mosaic virus. FEBS Lett 359:247-250
Tackaberry ES, Dudani AK, Prior F, Tocchi M, Sardana R, Altosaar I, Ganz PR (1999) Development of biopharmaceuticals in plant expression systems: cloning, expression and immunological rectivity of human cytomegalovirus glycoprotein B (UL55) in seeds of transgenic tobacco.
Vaccine 17:3020-3029
Tacket CO, Mason HS, Losonsky G, Clements JD, Levine MM, Arntzen JC (1998) Immunogenicity
in humans of a recombinant bacterial-antigen delivered in transgenic potato. Nat Med 4:607-609
Tacket CO, Mason HS, Losonsky G, Estes MK, Levine MM, Arntzen CJ (2000) Human immune
responses to a novel norwalk virus vaccine delivered in transgenic potatoes. J Infect Dis
182:302-305
Terashima M, Murai Y, Kawamura M, Nakanishi S, Stoltz T, Chen L, Drohan W, Rodriguez RL,
Katoh S (1999) Production of functional human alpha 1-antitrypsin by plant cell culture. Appl
Microbiol Biotechnol 52:516-523

592

Lico-Benvenuto-4

26-10-2005

9:26

Pagina 593

LICO ET AL.

Thanavala Y, Yang Y, Lyons P, Mason HS, Arntzen C (1995) Immunogenicity of transgenic


plant-derived hepatitis B surface antigen. Proc Natl Acad Sci USA 92:3358-3361
Tinland B (1996) The integration of T-DNA into plant genome. Trends Plant Sci 1:178-184
Toth RL, Chapman S, Carr F, Santa Cruz S (2001) A novel strategy for the expression of foreign
genes from plant virus vectors. FEBS Lett 489:215-219
Tuboly T, Yu W, Bailey A, Degrandis S, Du S, Erickson L, Nagy E (2000) Immunogenicity of
porcine transmissible gastroenteritis virus spike protein expressed in plants. Vaccine
18:2023-2028
Turpen TH, Reinl SJ, Charoenvit Y, Hoffman SL, Fallarme V, Grill LK (1995) Malarial epitopes
expressed on the surface of recombinant tobacco mosaic virus. Bio/Technol 13:53-57
Vain P, Finer KR, Engler DE, Pratt RC, Finer JJ (1996) Intron-mediated enhancement of gene
expression in maize (Zea mays L.) and bluegrass (Poa pratensis L.). Plant Cell Rep 15:489-494
Valdes R, Gomez L, Padilla S, Brito J, Reyes B, Alvarez T, Mendoza O, Herrera O, Ferro W, Pujol
M, Leal V, Linares M, Hevia Y, Garcia C, Mila L, Garcia O, Sanchez R, Acosta A, Geada D,
Paez R, Luis Vega J, Borroto C (2003) Large-scale purification of an antibody directed against
hepatitis B surface antigen from transgenic tobacco plants. Biochem Bioph Res Co 308:94-100
Vaquero C, Sack M, Chandler J, Drossard J, Schuster F, Schillberg S, Fischer R (1999) Transient
expression of a tumor-specific single chain fragment and a chimeric antibody in tobacco leaves.
Proc Natl Acad Sci USA 96:11128-11133
Vaquero C, Sack M, Schuster F, Finnern R, Drossard J, Schumann D, Reimann A, Fischer R (2002)
A carcinoembryonic antigen-specific diabody produced in tobacco. FASEB J 16:408-410
Verch T, Yusibov V, Koprowski H (1998) Expression and assembly of a full-length monoclonal antibody in plants using a plant virus vector. J Immunol Methods 220:69-75
Verch T, Hooper DC, Kiyatkin A, Steplewski Z, Koprowski H (2004) Immunization with a
plant-produced colorectal cancer antigen. Cancer Immunol Immun 53:92-99
Walmsley AM, Arntzen CJ (2003) Plant cell factories and mucosal vaccine. Curr Opin Biotech
14:145-150
Witcher DR, Hood EE, Peterson D, Bailey M, Bond D, Kusnadi A, Evangelista R, Nikolov Z,
Wooge C, Mehigh RJ, Kappel WK, Register J, Howard JA (1998) Commercial production of
-glucuronidase (GUS): a model system for the production of proteins in plants. Mol Breed
4:301-312
Yusibov V, Modelska A, Steplewski K, Agadjanyan M, Weiner D, Hooper DC, Koprowsky H
(1997) Antigens produced in plants by infection with chimeric plant viruses immunize against
rabies virus and HIV-1. Proc Natl Acad Sci USA 94:5784-5788
Yusibov V, Hooper DC, Spitsin SV, Fleysh N, Kean RB, Mikheeva T, Deka D, Karasev A, Cox S,
Randall J, Koprowski H (2002) Expression in plants and immunogenicity of plant virus-based
experimental rabies vaccine. Vaccine 20:3155-3164
Zeitlin L, Olmsted SS, Moench TR, Co MS, Martinell BJ, Paradkar VM, Russell DR, Queen C,
Cone RA, Whaley KJ (1998) A humanized monoclonal antibody produced in transgenic plants
for immunoprotection of the vagina against genital herpes. Nat Biotechnol 16:1361-1364
Zhang B, Yang YH, Lin YM, Rao Q, Zheng GG, Wu KF (2003) Expression and production of
bioactive human interleukin-18 in transgenic tobacco plants. Biotechnol Lett 25:1629-1635
Zhu Z, Hughes K, Huang L, Sun B, Liu C, Li Y (1994) Expression of human alpha-interferon in
plants. Virology 172:213-222
Zuo J, Chua N-H (2000) Chemical-inducible systems for regulated expression of plant genes. Curr
Opin Biotechn 11:146-151

593

Você também pode gostar