Sie sind auf Seite 1von 7

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

net/publication/7091362

Expression of a human anti-rabies virus monoclonal antibody in tobacco cell


culture

Article  in  Biochemical and Biophysical Research Communications · July 2006


DOI: 10.1016/j.bbrc.2006.03.219 · Source: PubMed

CITATIONS READS

49 170

6 authors, including:

Marzena J Fabis-Pedrini V. Pétiard


Perron Institute for Neurological and Translational Science Plant Sciences Integrator
52 PUBLICATIONS   795 CITATIONS    81 PUBLICATIONS   3,148 CITATIONS   

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Marzena J Fabis-Pedrini on 09 November 2018.

The user has requested enhancement of the downloaded file.


Biochemical and Biophysical Research Communications 345 (2006) 602–607
BBRC
www.elsevier.com/locate/ybbrc

Expression of a human anti-rabies virus monoclonal antibody


in tobacco cell culture q
Loı̈c Stéphane Girard a, Marzena Jolanta Fabis a, Maryse Bastin b, Didier Courtois b,
Vincent Pétiard b,*, Hilary Koprowski a,*
a
Thomas Jefferson University, 1020 Locust Street, Biotechnology Foundation, Philadelphia, PA 19107, USA
b
Centre de Recherche Nestlé-Tours, 101 Avenue G. Eiffel, BP 49716, 37097 Tours Cedex 2, France

Received 15 March 2006


Available online 19 April 2006

Abstract

A Nicotiana tabacum cv. Xanthi cell culture was initiated from a transgenic plant expressing a human anti-rabies virus monoclonal
antibody. Within 3 months, plant cell suspension cultures were established and recombinant protein expression was examined. The anti-
body was stably produced during culture growth. ELISA, protein G purification, Western blotting, and neutralization assay confirmed
that the antibody was fully processed, with association of light and heavy-chains, and that it was able to bind and neutralize rabies virus.
Quantification of antibody production in plant cell suspension culture revealed 30 lg/g of cell dry weight for the highest-producing cul-
ture (0.5 mg/L), 3 times higher than from the original transgenic plant. The same production level was observed 3 months after cell cul-
ture initiation. Plant cell suspension cultures were successfully grown in a new disposable plastic bioreactor, with a growth rate and
production level similar to that of cultures in Erlenmeyer flasks.
Ó 2006 Elsevier Inc. All rights reserved.

Keywords: Antibody; Disposable plastic bioreactors; Plant cell culture; Rabies; Recombinant; Xanthi

Plant-based systems are increasingly used for the pro- nant protein production [5,6]. Protein stability during
duction of recombinant proteins, because of several advan- production and storage is an important determinant of
tages over other production systems, such as the ability to the production cost. Indeed, degradation by proteases
carry out necessary post-translational modifications not can be avoided by the addition of a ‘‘Lys-Asp-Glu-Leu’’
available in bacterial systems, as well as greater safety (KDEL) signal to the protein sequence, which leads to
and lower production costs than those provided by ani- storage of the heterologous protein inside the endoplasmic
mal-based systems. reticulum (ER). This ER targeting also results in a
Plant-based technology has recently been extensively glycosylation pattern closer to the human one, by prevent-
reviewed, with a full description of plants commonly used, ing addition of plant-specific glycans, fucose, and xylose
a listing of proteins expressed thus far [1–5], and the poten- to the protein [7] thus avoiding a potential source of
tial economic superiority of these systems for the recombi- unwanted immunogenicity in the recombinant protein
while injected [8].
Many proteins relevant in human disease treatment have
q
Abbreviations: cv., cultivar; DW, dry weight; ER, endoplasmic already been expressed in plant-based systems (reviewed in
reticulum; FW, fresh weight; HC, heavy-chain; LC, light-chain; mAb, [2,9]), mainly in the form of edible vaccines [10,11] and
monoclonal antibody. monoclonal antibodies (mAb) [5,12]. A safe and cost-effec-
*
Corresponding authors. Fax: +33 0 2 47 49 14 14 (V. Pétiard); +1 215
923 6795 (H. Koprowski).
tive source of mAb is particularly desirable in the treatment
E-mail addresses: vincent.petiard@rdto.nestle.com (V. Pétiard), of human rabies virus infection, since current protocols rely
hilary.koprowski@jefferson.edu (H. Koprowski). heavily on anti-rabies immunoglobulins prepared mainly

0006-291X/$ - see front matter Ó 2006 Elsevier Inc. All rights reserved.
doi:10.1016/j.bbrc.2006.03.219
L.S. Girard et al. / Biochemical and Biophysical Research Communications 345 (2006) 602–607 603

from horse serum; not only are such immunoglobulins 1/10,000 dilution] conjugated to horseradish peroxidase (Jackson Immu-
expensive to prepare, accounting for their limited supply noResearch). Whole-plant-derived mAb was used as a positive control.
worldwide, but they present the risk of cross-contamina- Suspension culture in disposable bioreactors
tion with known or unknown horse pathogens [13]. Con-
sidering that rabies virus exposure causes an economic A new type of plastic bag-based bioreactor (Nestlé Research Center,
burden in the US of about $1 billion per year for vaccina- Tours) was used to scale-up the cell culture from 500 mL to 10 L. After
tion and immunoglobulin administration [14], a plant- sterilization of the disposable bioreactor, sterile medium was inserted into
the bioreactor and cells were inoculated [40 g/L (FW)].
based system of immunoglobulin production, as recently
developed in tobacco plants [15], is attractive. However, Small-scale purification of plant cell-derived antibodies
mAb expression levels in field plants vary depending on
plant maturation stage and climatic influences [16 and per- Lyophilized cells [100 g dry weight (DW), equivalent to 1.45 kg FW]
sonal observations], whereas plant cell cultures are based were mixed with 1.5 L of extraction buffer (20 mM sodium phosphate, pH
7.2) and disrupted using a French press. Soluble proteins were recovered
on undifferentiated cells growing in a fully controlled envi- by centrifugation (6000g, 20 min, 4 °C) and filtered on fiberglass (from
ronment, ensuring a constant level of expression and pre- 2.7- to 0.22-lm pore sizes). Cross-filtration on a 30-kDa membrane was
venting contamination of the environment. used to concentrate the supernatant to 300 mL. Soluble protein extracts
Here, we demonstrate for the first time the expression of were purified with an Äkta purifier equipped with a HiTrapä protein G
a human anti-rabies virus glycoprotein mAb in plant cell high-performance affinity column (Amersham Biosciences) according
to the manufacturer’s recommendations. mAb was eluted with 0.1 M
culture. A low-cost biomass scale-up was performed with glycine–HCl (pH 2.7) and pH was neutralized with 1 M Tris–HCl (pH
a new disposable bioreactor. 9.0).

Materials and methods ELISA

Plant material Binding of transgenic mAb to rabies virus was assessed by ELISA as
described [18], with the following modifications: 3 lg/mL of ERA rabies
A stable (T3) transgenic tobacco plant (Nicotiana tabacum cv. Xanthi) virus was used to coat the plates, followed by loading with 95 lL of serial
expressing an anti-rabies virus mAb [15] was used and is designated herein 3-fold dilutions of soluble extracted proteins obtained from 7 mg of
as R12. Briefly, the heavy-chain (HC) sequence, fused with a sequence lyophilized plant cell extract disrupted in 1 mL of sodium phosphate
encoding the KDEL ER retention signal, is placed under control of the extraction buffer. Human anti-rabies virus mAb (1 lg/mL) was used as a
cauliflower mosaic virus 35S promoter with duplicated upstream B positive control. Recombinant bound mAb were detected by successive
domains (Ca2p); the light-chain (LC) sequence is under the control of the additions of biotin-conjugated anti-human IgG1 (1:1000; BD PharMin-
potato proteinase inhibitor II promoter (Pin2p) [15]. gen, San Diego, CA), avidin alkaline phosphatase-conjugated antibody
A well-established wild-type BY2 tobacco cell suspension (N. tabacum) (Sigma), and p-nitrophenyl phosphate (Sigma) as substrate. Absorbance
was used for comparison to newly established tobacco cell suspensions. was read at 405 nm.

Plant cell culture In vitro neutralization assay

Callus induction. Primary explants were cut from 3-week-old wild-type Neutralizing activity of purified antibodies was assessed using a stan-
(WT) or R12 transgenic seedlings and placed on Murashige and Skoog- dard 20-h rapid fluorescent focus inhibition test (RFFIT) as described [18].
based medium, supplemented with 1 mg/L naphthalene acetic acid Briefly, diluted purified mAb produced by plant cell culture was incubated
(NAA), 0.1 mg/L kinetin, 30 g/L sucrose, and 8 g/L agarose (MAK), with rabies virus (CVS-11, CVS-N2C or ERA). Baby hamster kidney
under photoperiod conditions (16 h light/8 h dark), at 25 °C. The medium (BHK) cells were added to plates, incubated, and fixed. FITC-anti-rabies
was autoclaved before explants for 20 min at 115 °C. Resulting calli were nucleoprotein monoclonal globulin (Centocor, Malvern, PA) was added
subcultured monthly on Petri dishes. and plates were examined under a fluorescence microscope to detect
Initiation of suspension culture. Randomly picked calli (20 g) were infected cells. The original mAb produced in human hybridoma cells was
inoculated into 50 mL MAK liquid medium and placed under photope- used as positive control. Both mAb were used at the same concentration to
riod conditions at 25 °C on an orbital shaker (100 rpm). After 2 weeks, compare their neutralization ability.
both transformed and non-transformed tobacco cell lines were grown in
250-mL Erlenmeyer flasks containing 100 mL MAK medium and sub-
Results
cultured every 20 days at an initial density of 40 g of cells [fresh weight
(FW)]/L.
Morphological features of plant cell culture
Protein extraction and analysis
Primary calli initiated from WT tobacco seedlings and
Protein extraction, SDS–PAGE, and Western blotting were carried out R12 transgenic seedlings expressing human anti-rabies
as described [17], with the following modifications. Twenty-day suspension
cultures were filtered (50-lm nylon mesh) to separate cells and medium. virus mAb were grown on MAK medium and after 3 weeks
Cell disruption was performed on 1 g FW of cells, using a stainless-steel appear morphologically similar (Fig. 1A). The yellow color
mixer mill (Retsch, Model MM301, Newtown, PA) at 30 hits/s, for 2 min. turned to green after 3 months. Western blot analysis of
The resulting mash was homogenized with 300 lL DB2X buffer (40 mM eight WT and eight R12 calli revealed HC and LC protein
Tris–HCl, pH 8.0, 20% glycerol, 2% SDS, and 4% b-mercaptoethanol) production only in the R12 calli (data not shown).
containing 0.1% Triton X-100 and a complete protease inhibitor mixture
(Roche Diagnostics). Three weeks after inoculation, WT and R12 suspension
For Western blot analysis, HC and LC were detected with goat cultures appeared healthy in MAK medium (Fig. 1B), with
anti-human antibodies [Fc c- and F(ab 0 )2 fragment specific, respectively, the same yellow color and the same morphology as a stabi-
604 L.S. Girard et al. / Biochemical and Biophysical Research Communications 345 (2006) 602–607

Fig. 1. Morphological features of calli and plant cell suspension cultures grown in MAK medium. (A) Three-week-old calli, obtained by callogenesis on
Nicotiana tabacum (cv. Xanthi) plant leaves grown on MAK medium (left, wild-type calli; right, R12 calli). (B) Comparison of wild-type (left) and R12
transformed (right) 10-day-old Nicotiana tabacum cv. Xanthi suspension cell culture. (C) Filtered cells from 20-day-old plant cell suspension. Note the
greenish color and clumped growth.

lized N. tabacum BY2 culture. One month later, cells began extracts (Fig. 2B, lanes 1 and 3), loaded at a similar
to grow as clumps and turned greenish (Fig. 1C); cell sus- protein concentration as shown by SDS–PAGE and
pension cultures retained these features during further Coomassie blue staining (Fig. 2A). Analysis of four succes-
subcultures. sive extractions from the R12 remaining pellet revealed the
presence of HC and LC bands (data not shown). Expres-
Heavy- and light-chain expression in plant suspension sion of heterologous proteins was observed in all R12
cultures and pellets suspensions.

Western blot analysis (Fig. 2B) of soluble protein cell Increased monoclonal antibody production in plant cell
extracts from five R12 suspension cultures and their suspension culture
remaining pellets (after a second extraction) revealed HC
and LC bands migrating at 55 and 30 kDa, respectively. ELISA results for mAb expression in the transgenic
The LC appeared to be expressed as two different forms plant [15] indicated levels at 3 lg/g leaf FW, corresponding
(Fig. 2B, lanes 2 and 4). Two other bands were also to 9 lg/g leaf DW (assuming 70% water content in
detected in the pellet at 60 and 75 kDa (Fig. 2B). The leaves), whereas similar analysis in plant cell suspensions
60-kDa band appears to be a dimer of LC since this band using lyophilized cells indicated 10 lg/g DW for four sus-
was not detected by HC-specific antibody (data not pension cell cultures and 30 lg/g DW for the highest-yield
shown). The 75-kDa band might be an LC–HC dimer culture.
resulting from partial denaturation of the mAb. None of
these specific bands was detected in WT soluble protein In vitro neutralizing activity of monoclonal antibodies

The neutralization activity of mAb produced by plant


cell cultures was as efficient as the mAb produced by
hybridoma cells in a standard neutralization assay against
cell culture-adapted rabies virus strains (CVS-11, CVS-
N2C, and ERA). The same quantity of each mAb was
required to neutralize virus.

Kinetics of plant cell growth and monoclonal antibody


expression

Both WT and R12 cell suspensions were grown for 30


Fig. 2. Analysis of protein extracts of WT and R12 cells by SDS–PAGE days and examined every 4 days for fresh weight (data
stained with Coomassie blue (A) and Western blotting to detect human not shown) and dry weight (Fig. 3A). A well-established
anti-rabies virus monoclonal antibody light- and heavy-chains (B). Lanes
1 and 2 correspond to soluble protein extracts of wild-type and R12 cells
N. tabacum BY2 suspension culture was used for compar-
from suspension cell cultures, respectively; lanes 3 and 4 correspond to ison. WT and R12 cell cultures showed similar growth
proteins extracted from wild-type and R12 remaining pellets, respectively. curves, with no exponential phase, starting from 4.5 g/L
L.S. Girard et al. / Biochemical and Biophysical Research Communications 345 (2006) 602–607 605

Fig. 4. Western blot analysis of human anti-rabies virus monoclonal


antibody light- and heavy-chains in tobacco cell extracts after protein G
purification. FT, flowthrough; W, washing step; ELU, elution step; T+,
positive control (mAbP purified from plant on protein A column).

Fig. 3. Monoclonal antibody production in plant cell cultures grown in


mAb purified from whole plants (Fig. 4). From 100 g
Erlenmeyer flasks. (A) Growth curve of wild-type (-h-) and R12 (-j-) DW of cells, 250 lg of mAb was collected, which represents
Nicotiana tabacum cv. Xanthi, and wild-type Nicotiana tabacum BY2 (-d-). 25% of the measured production.
Data are the average of four samples and error bars correspond to the
resulting standard deviation. (B) Western blot detection of human anti- Discussion
rabies virus LC expressed in R12 cells with respect to growth curve.

In an effort to increase the safety and reproducibility of


and reaching 18 g/L final DW in 20 days. The BY2 cell cul- human anti-rabies virus mAb production in plant-based
ture reached 14 g/L in 10 days, starting from 0.6 g/L. systems, a transgenic tobacco plant expressing this recom-
Western blot analysis of HC (data not shown) and LC binant protein was grown as an undifferentiated cell cul-
(Fig. 3B) expression in R12 cells revealed the same level ture. WT and R12 calli were rapidly obtained on MAK
of expression at all time points examined. ELISA of extra- medium, and LC and HC expression was readily detected
cellular medium precipitated with 70% ammonium sul- in R12 calli by Western blotting. All calli were morpholog-
phate showed no significant difference between WT and ically healthy and identical, indicating that mAb expression
R12 suspension culture with respect to the presence of does not adversely affect cell metabolism. Moreover, LC
mAb chains, and Western blot revealed only the LC in and HC expression proved that both the Ca2p and Pin2p
the R12 medium, as a monomer (30 kDa) and a dimer promoters are also functional in undifferentiated plant
(60 kDa). cells.
Six weeks later, plant cell cultures were initiated in
Bioreactor scale-up of monoclonal antibody production MAK medium and examined for mAb production. Expres-
sion of the mAb appeared to remain constant throughout
Cells were grown for 15 days in Erlenmeyer flasks or a the growth periods, with no effect on cell growth rate.
disposable plastic bioreactor and harvested for analysis However, the presence of clumps, the lack of an exponen-
of biomass and mAb expression. In both systems, biomass tial growth phase, and the slow growth rate indicated the
production was on average of 13 g/L DW. mAb expression need for improved growth conditions. Indeed, N. tabacum
as determined using ELISA was also comparable, with BY2 cells are about 10 times more efficient than Xanthi
10 lg/g DW in flasks and 13 lg/g DW in the bioreactor. cells for biomass production, reflecting the decades of opti-
mization of BY2 cell growth conditions while Xanthi cells
Small-scale purification of plant-produced monoclonal were rarely used. Despite the relatively low growth rate of
antibodies Xanthi cell cultures, a biomass scale-up was performed
with a new disposable plastic bioreactor and showed simi-
R12-produced mAb was purified on a protein G col- lar growth rate and mAb expression levels to those in
umn, and flowthrough, washing step, and elution fractions Erlenmeyer flask cultures. Disposable bioreactors up to a
were examined for the presence of the HC and LC by Wes- size of 150 L for biomass production of BY2 cell culture
tern blotting (Fig. 4). WT cells were used as negative con- also showed similar results (personal communication).
trol. No HC or LC was detected in the WT sample or in the These data demonstrate that inexpensive and easy-to-use
flowthrough or the washing step phases of the R12 extract, disposable bioreactors are suitable for biomass production
confirming the high specificity of the column. Both LC and and for the production of recombinant proteins.
HC were detectable in the elution phase, in the same range Analysis of the mAb concentration from plant cell sus-
of intensity, and at the same molecular weight position as pension cultures by ELISA revealed a production level as
606 L.S. Girard et al. / Biochemical and Biophysical Research Communications 345 (2006) 602–607

high as and even higher than that in whole transgenic Acknowledgments


plants (up to 30 lg/g DW compared to 9 lg/g DW, respec-
tively). The production level in plant cells is probably high- We thank Dr. K. Ko for providing seedlings and mAb
er than 30 lg/g DW since several successive extractions controls, and Dr. D.C. Hooper for fruitful discussion.
revealed some mAb remaining in the pellet, probably We also thank C. Brimer, R. Kean, P. Hilaire, and B.
trapped in the ER. Since the ELISA used is based on rabies Terrier for technical assistance. This work was supported
virus recognition, the plant cell-produced mAb was pre- by a grant from USDA to Biotechnology Foundation (to
sumably fully processed, i.e., with LC and HC association, H.K.).
and functional, i.e., able to bind the virus. This resulting
mAb was also as effective as human hybridoma cell-pro- References
duced mAb in neutralizing three different rabies virus
strains, preventing infection of BHK cells. This result [1] S. Hellwig, J. Drossard, R.M. Twyman, R. Fischer, Plant cell cultures
showed that undifferentiated plant cells were able to pro- for the production of recombinant proteins, Nat. Biotechnol. 22
(2004) 1415–1422.
duce mAb with the same neutralization activity as whole
[2] N.P. Teli, M.P. Timko, Recent developments in the use of transgenic
transgenic plants, also compared to human hybridoma plants for the production of human therapeutics and biopharmaceu-
cell-produced mAb [15]. ticals, Plant Cell Tiss. Org. 79 (2004) 125–145.
The glycosylation pattern of the mAb and several other [3] T.J. Menkhaus, Y. Bai, C. Zhang, Z.L. Nikolov, C.E. Glatz,
characteristics, such as immunogenicity, remain to be Considerations for the recovery of recombinant proteins from plants,
examined. However, a high mannose-glycosylation like Biotechnol. Prog. 20 (2004) 1001–1014.
[4] S. Schillberg, R.M. Twyman, R. Fischer, Opportunities for recombi-
that for whole-plant-produced mAb is expected due to nant antigen and antibody expression in transgenic plants—technol-
the KDEL ER-retention signal [7,8]. This hypothesis is ogy assessment, Vaccine 23 (2005) 1764–1769.
supported by the presence in Western blots of both LC [5] E. Stoger, M. Sack, L. Nicholson, R. Fischer, P. Christou, Recent
and HC at the same molecular weight (30 and 55 kDa, progress in plantibody technology, Curr. Pharm. Des. 11 (2005) 2439–
respectively) as the glycosylated form of mAb produced 2457.
[6] S. Nandi, D. Yalda, S. Lu, Z. Nikolov, R. Misaki, K. Fujiyama, N.
by the whole plant [8]. The LC might present two glycosyl- Huang, Process development and economic evaluation of recombi-
ation patterns since a second band was observed. The nant human lactoferrin expressed in rice grain, Transgenic Res. 14
KDEL signal expressed on HC also prevented mAb secre- (2005) 237–249.
tion into the medium. The small amount of mAb detected [7] A. Triguero, G. Cabrera, J.A. Cremata, C.T. Yuen, J. Wheeler, N.I.
Ramirez, Plant-derived mouse IgG monoclonal antibody fused to
in the R12 medium might be released by dying cells. Only
KDEL endoplasmic reticulum–retention signal is N-glycosylated
LC was detected outside of the cells, probably secreted via homogeneously throughout the plant with mostly high-mannose-type
the ‘‘default pathway’’ when not bound to HC. N-glycans, Plant Biotechnol. J. 3 (2005) 449.
The amount of human anti-rabies virus mAb recovered [8] Y. Tekoah, K. Ko, H. Koprowski, D.J. Harvey, M.R. Wormald,
from 100 g DW of R12 cells was 4-fold less than in pilot R.A. Dwek, P. Rudd, Controlled glycosylation of therapeutic
experiments, probably due to mAb precipitation during antibodies in plants, Arch. Biochem. Biophys. 426 (2004) 266–278.
[9] J.K. Ma, E. Barros, R. Bock, P. Christou, P.J. Dale, P.J. Dix, R.
the long extraction process [17]. In the present study, the Fischer, J. Irwin, R. Mahoney, M. Pezzotti, S. Schillberg, P. Sparrow,
main compounds responsible for mAb precipitation are E. Stoger, R.M. Twyman, Molecular farming for new drugs and
likely the polyphenols, as described by Haslam [19]. These vaccines—current perspectives on the production of pharmaceuticals
compounds form aggregates, possibly corresponding to the in transgenic plants, EMBO Rep. 6 (2005) 593–599.
dark precipitates observed in the supernatant even within a [10] H. Koprowski, Vaccines and sera through plant biotechnology,
Vaccine 23 (2005) 1757–1763.
few minutes after filtration through 0.22-lm filters. These [11] S.J. Streatfield, Plant-based vaccines for animal health, Rev. Sci.
compounds imposed multiple filtration steps, several times Tech. 24 (2005) 189–199.
during the purification, increasing the processing time to [12] R. Valdes, L. Gomez, S. Padilla, J. Brito, B. Reyes, T. Alvarez, O.
4 h. Another phenomenon has been described [20,21], Mendoza, O. Herrera, W. Ferro, M. Pujol, V. Leal, M. Linares, Y.
Hevia, C. Garcia, L. Mila, O. Garcia, R. Sanchez, A. Acosta, D.
whereby 80% of a mAb dissolved in an Erlenmeyer flask
Geada, R. Paez, J. Luis Vega, C. Borroto, Large-scale purification of
with pure Murashige and Skoog-medium can ‘‘disappear’’ an antibody directed against hepatitis B surface antigen from
in less than 2 h by anchoring onto the glassware wall. Such transgenic tobacco plants, Biochem. Biophys. Res. Commun. 308
adsorption of the mAb might also explain the low recovery. (2003) 94–100.
Further studies are needed to overcome these obstacles to [13] WHO, Consultation on a monoclonal antibody cocktail for rabies
optimal mAb recovery. post-exposure treatment. www.who.int/rabies/vaccines/en/mabs_fi-
nal_report.pdf Geneva, 23–24 May 2002 (2002) 1–8.
Plant cell culture is a developing technology for recom- [14] ACIP, Recommendations of the Advisory Committee on Immuniza-
binant protein production and presents great potential in tion Practices. Centers for Disease Control and Prevention. Human
competing with other production systems. Many features rabies prevention-United States, Morbidity & Mortality Weekly Rep.
remain to be optimized such as the expression level of the 48 (1999) 1–21.
[15] K. Ko, Y. Tekoah, P.M. Rudd, D.J. Harvey, R.A. Dwek, S. Spitsin,
product and the purification processes. Currently, the plant
C.A. Hanlon, C. Rupprecht, B. Dietzschold, M. Golovkin, H.
cell culture system is more suitable for the production of Koprowski, Function and glycosylation of plant-derived antiviral
recombinant proteins needed in small quantity and pre- monoclonal antibody, Proc. Natl. Acad. Sci. USA 100 (2003) 8013–
senting high activity. 8018.
L.S. Girard et al. / Biochemical and Biophysical Research Communications 345 (2006) 602–607 607

[16] L.H. Stevens, G.M. Stoopen, I.J. Elbers, J.W. Molthoff, H.A. Bakker, substitute for pooled human immune globulin in the prophylactic
A. Lommen, D. Bosch, W. Jordi, Effect of climate conditions and treatment of rabies virus exposure, J. Immunol. Methods 235
plant developmental stage on the stability of antibodies expressed in (2000) 81–90.
transgenic tobacco, Plant Physiol. 124 (2000) 173–182. [19] E. Haslam, Plant Polyphenols—Vegetable Tannins Revisited, Cam-
[17] L.S. Girard, M. Bastin, D. Courtois, Expression of the human milk bridge University Press, Cambridge, 1989, pp. 167–192.
protein sCD14 in tobacco cell culture, Plant Cell Tiss. Org. 78 (2004) [20] B.M. Tsoi, P.M. Doran, Effect of medium properties and additives on
253–260. antibody stability and accumulation in suspended plant cell cultures,
[18] J.M. Champion, R.B. Kean, C.E. Rupprecht, A.L. Notkins, H. Biotechnol. Appl. Biochem. 35 (2002) 171–180.
Koprowski, B. Dietzschold, D.C. Hooper, The development of [21] P.M. Doran, Loss of secreted antibody from transgenic plant tissue
monoclonal human rabies virus-neutralizing antibodies as a cultures due to surface adsorption, J. Biotechnol. 122 (2006) 39–54.

View publication stats

Das könnte Ihnen auch gefallen