You are on page 1of 11

Journal of Immunological Methods 296 (2005) 199 – 209

www.elsevier.com/locate/jim

Research paper

Microencapsulation for the gastric passage and controlled


intestinal release of immunoglobulin Y
Jennifer Kovacs-Nolan, Yoshinori Mine*
Department of Food Science, University of Guelph, Guelph, Ontario, Canada N1G 2W1
Received 11 August 2004; received in revised form 12 November 2004; accepted 16 November 2004
Available online 16 December 2004

Abstract

Avian immunoglobulin (Ig) Y is a promising alternative for the treatment and prevention of enteric infections, and has
shown to be effective against a number of gastrointestinal pathogens, including Escherichia coli, Salmonella, and rotavirus.
However, its application is limited by its sensitivity to human gastrointestinal conditions. Here, we report on the enteric coating
of IgY-containing granules with a pH-sensitive methacrylic acid copolymer. A 35% (w/w) polymer coating was found to protect
IgY from gastric inactivation in vitro, maintaining greater than 95% activity after 6 h in simulated gastric fluid. The IgY was
slowly released from the microencapsulated granules upon exposure to simulated intestinal fluid, and retained 80% activity after
8 h exposure to pancreatic enzymes. In vivo, using pigs as a model of human digestion, the encapsulated IgY retained
significantly more activity than non-encapsulated IgY, indicating that microencapsulation with a methacrylic acid copolymer
may be an effective method of protecting IgY from gastrointestinal inactivation, enabling its use for oral passive
immunotherapy.
D 2004 Elsevier B.V. All rights reserved.

Keywords: Immunoglobulin Y; Gastro-resistance; Methacrylic acid copolymer; Human rotavirus; VP8; Passive immunotherapy

1. Introduction

Passive immunization may be one of the most


valuable applications of antibodies (Sim et al., 2000).
Abbreviations: BSA, bovine serum albumin; ELISA, enzyme-
linked immunosorbent assay; GI, gastrointestinal; HRP, horseradish Oral immunotherapy, using pre-formed pathogen-
peroxidase; HRV, human rotavirus; Ig, immunoglobulin; MWCO, specific antibodies, has been examined as a method
molecular weight cut-off; PBS, phosphate-buffered saline; PEG, of establishing passive immunity against enteric
polyethylene glycol; SGF, simulated gastric fluid; SIF, simulated pathogens (Carlander et al., 2000), and has the
intestinal fluid; TBS, Tris-buffered saline; TBST, Tris-buffered advantage of reduced cost, ease of administration,
saline containing Tween-20.
* Corresponding author. Tel.: +1 519 824 4120x52901; fax: +1
and the potential to treat localized conditions in the
519 824 6631. gastrointestinal tract (Reilly et al., 1997). However, in
E-mail address: ymine@uoguelph.ca (Y. Mine). order to be effective, the antibodies must survive the
0022-1759/$ - see front matter D 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.jim.2004.11.017
200 J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209

gastrointestinal environment and reach their target tiveness, in some cases destroying antibody activity by
areas with their biological properties intact (Bogstedt the encapsulation procedure itself. The macroencap-
et al., 1997). sulation of IgY, using enteric-coated gelatin capsules,
Avian immunoglobulin (Ig) Y, the functional has also been examined, and was found to significantly
equivalent of IgG found in mammals, is ideal for improve antibody stability (Akita and Nakai, 2000).
passive immunotherapy applications, as it may be The anionic methacrylic acid–ethyl acrylate
readily obtained in large quantities from egg yolk, and copolymer, EudragitR L100-55, is a commonly used
presents a more cost-effective, convenient, and pharmaceutical coating. It is insoluble in acid medium,
hygienic alternative to mammalian antibodies (Car- but will begin to dissolve above pH 5.5. Coating with
lander et al., 2000). IgY has been shown to be this enteric polymer results in products which are
effective against a number of enteric pathogens, resistant to gastric conditions, and will dissolve readily
including Escherichia coli, Salmonella spp., and in the neutral to slightly alkaline environment of the
rotavirus, by preventing infection and inhibiting small intestine (Lehmann et al., 1999).
growth in vitro, and preventing or reducing symptoms Previously, we reported on the production of IgY
in animal models (Kovacs-Nolan and Mine, 2004). against the recombinant VP8 subunit of the human
However, to maximize the potential of IgY for rotavirus (HRV) (Kovacs-Nolan et al., 2001). HRV is
immunotherapeutic use in humans would require the major etiologic agent of acute infantile gastro-
protection of the IgY from acidic and enzymatic enteritis, infecting up to 90% of children under the age
degradation in the stomach, in order to reach the small of three (Kapikian and Chanock, 1996), and is
intestine, the site of infection, intact. characteristically localized to the epithelial cells of
While the survival of IgG has been reported in the gastrointestinal tract (Kapikian and Chanock,
individuals fed antibodies of human or bovine origin, 1996; Clark et al., 1999). Currently no effective
IgY is a much less stable molecule, owing to its higher vaccine exists. Passive immunization, to prevent and
molecular weight (180 kDa), lower amount of h-sheet treat rotavirus gastroenteritis, would require the
structure, and reduced flexibility (Warr et al., 1995; delivery of virus-neutralizing antibodies to the small
Shimizu et al., 1992), and therefore may be more intestine, the site of viral replication.
susceptible to degradation in the gastrointestinal tract. Here, we describe the microencapsulation of anti-
Although the stability of both immunoglobulins VP8 IgY using an anionic methacrylic acid–ethyl
has been found to be similar when subjected to acrylate copolymer (EudragitR L100-55), and the
alkaline conditions, IgY is denatured much more characterization and evaluation of the protective
readily than IgG in acidic conditions (Shimizu et al., efficacy of the enteric polymer coating under gastro-
1992, 1993a). IgY has been found to be relatively intestinal conditions.
resistant to trypsin and chymotrypsin digestion, but
sensitive to pepsin digestion. However, it is more
susceptible to pepsin, trypsin, and chymotrypsin 2. Materials and methods
digestion than IgG (Shimizu et al., 1993b). Hatta et
al. (1993) found that digestion of IgY with pepsin at 2.1. IgY production and separation
pH 2 resulted in complete hydrolysis of the antibody,
with a corresponding loss in activity, but activity Polyclonal antibodies (IgY) against recombinant
remained even after 8 h incubation with trypsin or HRV subunit protein, VP8, were produced in chick-
chymotrypsin. Therefore, an effective method of ens, as previously described (Kovacs-Nolan et al.,
protecting the IgY from pepsin degradation is 2001). Crude IgY was separated from the yolks of
necessary for the successful application of IgY to immunized hens using a modification of the method
treat and prevent enteric infections. of Akita and Nakai (1992). Briefly, egg yolks were
The microencapsulation of IgY using liposomes separated from the egg white, diluted 1:4 with
(Shimizu et al., 1993b) and multiple emulsions distilled water (which had been acidified with 0.1 M
(Shimizu and Nakane, 1995) have been previously HCl to give a final pH of 5–5.2 after dilution), and
described; however, these have shown limited effec- kept at 20 8C for 48 h. The frozen yolk solutions
J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209 201

were thawed at 4 8C, and centrifuged at 12 000g, for ratio of 1:250 (Eggum and Boisen, 1991), and
1 h at 10 8C, to precipitate lipoproteins (Jensenius et incubated, with shaking, at 37 8C. At intervals of
al., 1981). The resulting supernatant was then filtered 0, 0.5, 1, 2, 3, 4 and 6 h, samples were neutralized
and lyophilized. with 0.1 M sodium carbonate buffer, pH 9.6, and the
antibody activity was assessed by ELISA. Activities
2.2. Preparation of IgY granules were expressed as a percent relative to an untreated
control sample.
IgY granules were prepared by Snow Milk, Tokyo,
Japan. The crude IgY was first blended with corn 2.5. In vitro stability of IgY to simulated intestinal
starch (National Starch, Tokyo, Japan), at a ratio of 1 conditions
part IgY to 4 parts starch. Granules ranging in size
from 2 to 3 mm in diameter were formed using a Simulated intestinal fluid (SIF) was prepared as
Yokomizo Granular (Model FR160 60, Tokyo, described in the United States Pharmacopoeia (United
Japan), at a temperature of 32 8C, humidity of 78%, States Pharmacopeial Convention Council of Experts,
and rotation time of 5 min. 2004b), consisting of 10 mg/mL pancreatin (USP,
Sigma Chemical) in 0.05 M KH2PO4, at pH 6.8, and
2.3. Coating of the IgY granules with EudragitR was used to simulate small intestinal conditions. The
L100-55 encapsulated IgY was first incubated in SGF for 2 h.
The granules were then filtered and suspended in 0.05
The IgY granules were first sealed with a 5% (w/ M KH2PO4, pH 6.8, and SIF was added to give an
w) coating of PEG 6000 (USP, Fluka Chemie GmbH, enzyme to substrate ratio of 1:50 (Mouecoucou et al.,
Buchs, Switzerland), applied by immersing the 2004). Samples were incubated, with shaking, at 37
granules in a 10% (w/v) aqueous solution of PEG 8C. At intervals of 0, 0.5, 1, 2, 3, 4, 6, and 8 h,
6000, followed by air drying at 37 8C. samples were removed and placed on ice to stop the
A 15% (w/v) aqueous polymer dispersion was reaction. Non-encapsulated IgY was incubated in SIF
prepared according to the manufacturer’s instructions. only. The antibody activity was assessed by ELISA.
In short, EudragitR L100-55 (Rfhm GmbH, Darm- Activities were expressed as a percent relative to an
stadt, Germany) powder was added slowly to the untreated control sample.
appropriate volume of water, at room temperature,
with stirring, and 1 M NaOH was added to the polymer 2.6. In vivo gastrointestinal stability of IgY
solution to partially neutralize 5–15% of the carboxyl
groups. The PEG-coated granules were coated with the Six 28-day-old weaned Yorkshire pigs (weighing
polymer solution by repeated immersion in the 15% 6–7 kg) were obtained from Arkell Research Station,
polymer dispersion, followed by air drying at 37 8C, to Swine Unit (University of Guelph, Guelph, Ontario),
give final polymer coatings of 5%, 10%, 15%, 20%, and were kept in accordance with the Canadian
25%, 30%, 35%, and 40% (w/w). Council on Animal Care Guide to the Care and Use
of Experimental Animals. Water and food were
2.4. In vitro stability of IgY to simulated gastric provided ad libitum throughout the experiment, in
conditions order to maintain normal digestive functions.
Pigs were divided into two groups of three pigs
The stability of the encapsulated IgY to gastric each. Control pigs were each given 250 mg of non-
conditions was evaluated using simulated gastric encapsulated IgY, suspended in 30 mL of 0.05 M
fluid (SGF). SGF was prepared as described in the citrate buffer, pH 3.7, administered by oral gavage,
United States Pharmacopoeia (United States Pharma- using a 10-mL syringe attached to a length of tubing.
copeial Convention Council of Experts, 2004a), and Test pigs were given 2 g of encapsulated IgY granules
consisted of 3.2 mg/mL pepsin (Sigma Chemical, St. (equivalent to 250 mg of IgY), flushed down the
Louis, MO) in 0.03 M NaCl, at pH 1.2. The SGF tubing with 30 mL of 0.05 M citrate buffer, pH 3.7.
was added to the IgY to give an enzyme to substrate After 3 h, the pigs were euthanized, and necropsy was
202 J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209

immediately carried out to collect tissue samples from washed again, and 100 AL/well of horseradish pero-
the small intestine (duodenum, jejunum, and ileum) xidase (HRP)-conjugated anti-chicken IgG (Sigma
and large intestine, as well as stomach contents. Chemical), diluted 5000 in TBS containing 1% (w/
Tissue samples were added to 1–2 volumes of PBS, v) BSA, was added and incubated for 1 h, at 37 8C.
containing Completek protease inhibitor cocktail The plate was washed and developed using 100 AL/
tablets (Roche Diagnostics GmbH, Mannheim, Ger- well of 3,3V,5,5V-tetramethylbenzidine substrate sol-
many), and kept on ice. Stomach contents were added ution (TMB) (Sigma Chemical). The reaction was
to 3–6 volumes of protease inhibitor-containing PBS, stopped by the addition of 1 M H2SO4 (100 AL/well),
to neutralize stomach acid. and the absorbance at 450 nm was read using a BioRad
Samples were homogenized using a PolytronR PT model 550 plate reader.
2000 homogenizer (Kinematica AG, Littau-Luzern, For IgY quantification, plates were coated with 0.01
Switzerland) at 15 000 rpm, and centrifuged at Ag/well of rabbit anti-chicken IgY (Jackson Immu-
5500g for 10 min at 4 8C. Supernatants were passed noResearch Laboratories, West Grove, PA) in 0.1 M
through a 1.0-Am glass fiber filter to remove sodium carbonate buffer, pH 8.6. A standard curve was
particulate matter, and were then concentrated to 1/5 prepared using pure chicken IgY (Jackson ImmunoR-
of their original volume by ultrafiltration using a 50- esearch Laboratories), at concentrations from 0 to 32
kDa MWCO cellulose membrane (Millipore, Bedford, ng IgY/mL in TBS containing 1% (w/v) BSA.
MA). IgY activity determination and IgY quantifica- Detection was carried out as described above.
tion were done by ELISA, using standard curves of
anti-VP8 IgY and pure IgY, respectively. 2.9. SDS–PAGE

2.7. In vitro IgY release Sodium dodecyl sulfate polyacrylamide gel elec-
trophoresis (SDS–PAGE) was performed according to
The release of IgY from the encapsulated granules the method of Laemmli (1970). Samples were
was studied by incubating 100 mg of granules in 1 mL prepared under non-reducing conditions, and resolved
SGF, at 37 8C, with shaking. After 2 h, the granules using 10% acrylamide.
were filtered and transferred to 1 mL of 0.05 M
KH2PO4, pH 6.8, and incubated at 37 8C, with 2.10. Statistical analysis
shaking. At 0, 0.5, 1, 2, 3, 4, 6, and 8 h, 50-AL
aliquots were removed and assayed for protein Statistical analyses were performed using SPSS
concentration using the DC Assay (BioRad, Hercules, 7.5.2 for Windows, and statistical significance was
CA). The amount of protein released was expressed as determined using a paired-samples t-test.
a percent relative to the total protein in the sample.

2.8. IgY ELISA 3. Results

To assess IgY activity, 96-well microwell plates Initial coating experiments were carried out to
(Corning Costar, Cambridge, MA) were coated with optimize EudragitR L100-55 concentration and coat-
0.25 Ag/well of purified recombinant HRV subunit ing conditions for the anti-VP8 IgY granules. A first
protein, VP8, in 0.1 M sodium carbonate buffer, pH coat of PEG 6000, to seal the granules before polymer
9.6, and incubated at 4 8C overnight. The plate was application, was found to be necessary to improve the
washed with Tris-buffered saline (TBS) containing resistance of the coated granules to gastric conditions.
0.05% (v/v) Tween-20 (TBST), and blocked with 150 Polymer coatings of 5–40% (w/w) were examined for
AL/well of 2% (w/v) BSA (Fisher Scientific, Fair their ability to confer gastric stability to the IgY, and
Lawn, NJ) in 0.1 M sodium carbonate buffer, at 37 8C the results can be seen in Fig. 1. Initial samples were
for 2 h, with shaking. The plate was then washed with screened for IgY activity after 1 h in simulated gastric
TBST, and incubated at 37 8C, for 1 h, with 100 AL/ fluid (SGF). Antibody activity was defined as the
well of the IgY solution to be tested. The plate was ability of the anti-VP8 IgY to bind to recombinant
J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209 203

100
90
80
Relative activity (%)
70
60
50
40
30
20
10
0
PEG 5% 10% 15% 20% 25% 30% 35% 40%
% L100-55 (w/w)
Fig. 1. Optimization of methacrylic acid copolymer coating concentration. The ability of the different coating concentrations to protect anti-VP8
IgY against gastric inactivation was assessed by incubating samples for 1 h in SGF. Antibody activity was measured by ELISA, and expressed as
% activity relative to an untreated sample. Data are presented as meansFstandard errors (n=2).

VP8, and was measured by an antibody capture The stability of the IgY to small intestinal conditions
ELISA. The relative activities were expressed as a was examined using simulated intestinal fluid (SIF).
percentage of an untreated, positive control, sample IgY samples were first incubated in SGF, and then SIF,
(100% activity). A decrease in absorbance, as a result and monitored for antibody activity over a period of 8
of decreased antibody binding, was attributed to a loss h. Non-encapsulated IgY was incubated only in SIF,
of antibody activity. with no prior treatment in SGF. The percent of IgY
A minimum 35% (w/w) polymer coating was activity remaining after treatment was determined by
required for stability to gastric conditions, resulting ELISA (Fig. 3A). IgY activity in both non-encapsu-
in greater than 95% retention of IgY activity after 1 h lated and encapsulated samples was not significantly
in SGF. This polymer concentration was chosen for all affected ( pb0.005) by incubation in SIF, retaining
of the subsequent IgY microencapsulation experi- around 90% and 80% antibody activity, respectively,
ments. The microencapsulated IgY was tested in after 8 h. The microencapsulated IgY did not differ
parallel with non-encapsulated IgY to determine significantly ( pb0.005) from the non-encapsulated IgY
efficacy of the polymer coating under each of the in its susceptibility to protease digestion by trypsin and
conditions being tested. chymotrypsin, as both samples retained similar levels
Gastro-resistance, or stability of the IgY under of activity after 8 h of SIF exposure. The intact IgY was
simulated gastric conditions, was assessed by incubat- visualized by SDS–PAGE (Fig. 3B), and can be seen as
ing the IgY in SGF for 6 h. At each time interval, the a band around 180 kDa.
samples were neutralized to inhibit pepsin activity, The in vivo stability of the IgY was examined by
and IgY activity remaining was determined by ELISA feeding microencapsulated and non-encapsulated IgY
(Fig. 2A). After 30 min of SGF exposure, the non- to weaned pigs, and observing the progress of the IgY
encapsulated IgY demonstrated a significant decrease through the gastrointestinal tract. Three hours after
(86%) in IgY activity, which rapidly decreased to zero feeding of the IgY, samples were taken from several
following continued incubation. The microencapsu- sites along the gastrointestinal tract. The pH of the
lated IgY maintained greater than 95% activity after 6 samples was immediately measured following tissue
h in SGF. The corresponding intact IgY was visualized collection. The pH of the small and large intestine
by SDS–PAGE, and can be seen as a band around 180 samples ranged from 6 to 7; the stomach pH was
kDa (Fig. 2B). As this was a crude IgY preparation, around 2, and sufficient PBS was added to adjust to
additional smaller proteins present in the water soluble neutral pH. The IgY was extracted from the tissue
fraction, including other livetins, can also be seen. samples, and IgY activity was determined by an
204 J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209

A
100

Relative activity (%)


80

60

40

20

0
0 1 2 3 4 5 6
Time (hours)
B
kDa
205
IgY
116
97
84
66
55
45
36

mwm IgY 0h 1h 2h 3h 4h 6h
std

Fig. 2. In vitro stability of microencapsulated (4) and non-encapsulated (5) anti-VP8 IgY to simulated gastric conditions. (A) IgY activity
remaining over 6 h incubation in SGF was measured by ELISA, and expressed as % activity relative to an untreated sample. Data are presented
as meansFstandard errors (n=2). (B) Intact IgY was visualized by SDS–PAGE.

antibody capture ELISA. Because the IgY activity IgY per gram of tissue sample (Fig. 4), and indicated
observed in the samples from the gastrointestinal tract that a large amount of the IgY was still present in the
could not be expressed as a percentage relative to the stomach, although a small portion appeared to have
initial dose administered, the samples were instead traveled distally to the jejunum and ileum.
compared to a standard anti-VP8 IgY curve, of known In the case of the microencapsulated sample, there
dilutions, and reported as relative amounts of active appeared to be a positive correlation (r=0.996)
antibody, as determined from the curve. Throughout between the IgY activity and the amount of IgY in
the gastrointestinal tract, significantly more activity the different regions of the gastrointestinal tract. In
( pb0.05) was retained by the microencapsulated IgY contrast, the amount of non-encapsulated IgY in the
than the non-encapsulated IgY, which retained very gastrointestinal tract did not correlate as well with IgY
little activity (Fig. 4). The highest IgY activity activity (r=0.615), showing high quantities of IgY yet
appeared to be in the stomach, and, to a lesser extent, low IgY activity, suggesting that although the anti-
in the jejunum and ileum. bodies were present, they had lost their activity.
The in vivo gastrointestinal distribution, or transit, The sustained release characteristics of the encap-
of IgY in the gastrointestinal tract was visualized by sulated IgY granules were examined by incubating the
quantifying the IgY in each of the regions of the encapsulated IgY in SGF and subsequently in SIF, and
gastrointestinal tract. The results were expressed as ng measuring the percent of protein released over time
J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209 205

A
100

Relative activity (%)


80

60

40

20

0
0 0.5 1 2 3 4 5 6 8
Time (hours)
B
kDa
205
IgY

116
97
84
66

55
45
36

mwm IgY 0h 1h 2h 4h 6h 8h
std

Fig. 3. In vitro stability of microencapsulated ( ) and non-encapsulated (5) anti-VP8 IgY to simulated intestinal conditions. (A) IgY activity
remaining over 8 h incubation in SIF was measured by ELISA, and expressed as % activity relative to an untreated sample. Data are presented
as meansFstandard errors (n=2). (B) Intact IgY was visualized by SDS–PAGE.

(Fig. 5). After transfer to SIF, the granules began to difficult the use of orally administered antibodies for
disintegrate, releasing over 80% of protein within 2 h of the passive immunotherapy of enteric infections.
incubation in SIF. The granules continued to disinte- Studies of the passage of IgG, derived from bovine
grate, reaching 100% release after 4 h incubation. colostrum, through the gastrointestinal tract of
humans have demonstrated only 4–19% survival of
antibody activity (Hilpert et al., 1987; Roos et al.,
4. Discussion 1995; Pacyna et al., 2001). Bogstedt et al. (1997)
reported the detection of only a minute amount of
Ingested proteins are typically denatured by the antibodies, both IgG and IgY, following digestion.
acidic pH of the stomach, and degraded by the And Kelly et al. (1997), who investigated the survival
proteolytic enzymes present in the stomach and small of bovine antibodies against Clostridium difficile
intestine (Reilly et al., 1997), thereby rendering toxin, observed that loss of antibody activity follow-
206 J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209

4.50 45

4.00 40

IgY concentration (ng/g)


3.50 35
Relative activity (x10-4)

3.00 30

2.50 25

2.50 20

1.50 15

1.00 10

0.50 5

0.00 0
stomach duodenum jejunum ileum lg intestine
Region of GI tract
Fig. 4. In vivo stability of microencapsulated ( ) and non-encapsulated (5) anti-VP8 IgY. Anti-VP8 IgY activity in each region of the
gastrointestinal tract was assessed by ELISA, and expressed as the relative amount of IgY retaining activity. The amount of microencapsulated
(.) and non-encapsulated (E) IgY was determined by IgY quantification ELISA. Data are presented as meansFstandard errors (n=3).

ing passage through the human gastrointestinal tract stomach, and enable it to be released in the small
corresponded to a loss of toxin-neutralizing activity. intestine, the site of viral replication.
In order to passively prevent or treat enteric Pepsin, which is optimally active in acidic con-
infections, such as that caused by human rotavirus, ditions ranging from pH 1 to 3.5, is the predominant
the IgY must resist degradation in the stomach, and digestive enzyme in the stomach (Castro, 1981; Reilly
reach the small intestine with its activity intact. An et al., 1997). Transit time through the stomach is
effective method of IgY microencapsulation would highly variable, ranging from 0.5 to 4.5 h (Davis,
protect the IgY from the digestive conditions of the 1989a; Madsen, 1994) with an average time of 1 to

SGF SIF

100
90
Percent release (%)

80
70
60
50
40
30
20
10
0
0 1 2 3 4 5 6 7 8
Time (hours)
Fig. 5. Dissolution of encapsulated anti-VP8 IgY granules. Granules were first incubated in SGF, and then transferred to SIF, and the amount of
protein released over time was measured. Data are presented as meansFstandard errors (n=2).
J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209 207

1.5 h (Fallingborg et al., 1990; Madsen, 1994). The ditions had little effect on IgY activity, indicating that
activity of pepsin is terminated when the stomach the IgY remained unaffected by the digestive enzymes
contents are mixed with alkaline pancreatic juice in of the small intestine. As well, the activity of the
the small intestine. The principal enzymes present in encapsulated and non-encapsulated IgY did not differ
the small intestine are trypsin, chymotrypsin, elastase, significantly, suggesting that the structure of the IgY
and carboxypeptidase (Castro, 1981). Small intestinal was not altered by encapsulation, or by the prior
transit times typically range from 2 to 6 h, but it may exposure to SGF.
be as short as 1 h (Davis, 1989b). Several studies have been carried out to examine
A simulated gastric solution, mimicking conditions the survival of immunoglobulins of human or bovine
in the stomach, was used to assess the stability of IgY origin through the gastrointestinal tract (Bogstedt et
encapsulated with a pH-sensitive methacrylic acid al., 1997); however, less is known about the gastro-
copolymer (EudragitR L100-55) in vitro. Similar to intestinal fate of IgY. In order to assess the stability of
previous reports (Hatta et al., 1993), the non- the microencapsulated IgY in vitro, under physiolog-
encapsulated IgY was extremely sensitive to gastric ical conditions, an animal model was used.
conditions, and was rapidly inactivated. We have The anatomy, physiology, and biochemistry of the
demonstrated, however, that a 35% (w/w) enteric gastrointestinal tract differ considerably among mam-
polymer coating resulted in the retention of greater malian species. A number of physiological factors can
than 95% of IgY activity after 6 h incubation in SGF, modify dissolution rates and solubility, and transit
which is longer than the typical observed gastric times can be significantly different between species
transit time of humans. due to different dimensions and propulsive activities
Once the microencapsulated IgY was exposed to of the gastrointestinal tract. It is imperative, then, to
the simulated intestinal conditions (pHN5.5), the select the correct animal model to study gastro-
polymer coating gradually disintegrated, releasing intestinal function in the human (Kararli, 1995). Pigs
the IgY from the granules. The sustained release of are considered the best models for the study of
the IgY from the microencapsulated granules would digestion in humans, due to similar morphology and
ensure that, in vivo, the antibodies would be physiology of the gastrointestinal systems (Miller and
distributed slowly throughout the small intestine Ullrey, 1987; Rowan et al., 1994), and so were
during transit, available to bind and neutralize the utilized here to study the gastric stability of IgY.
target organism. Here, over 50% was released after the The microencapsulated IgY retained significantly
first hour, and 100% release from the encapsulated more activity than did the non-encapsulated IgY after
granules occurred between 3 and 4 h. The dissolution 3 h of digestion, indicating that the polymer is
profile of the granules, however, may be further capable of protecting IgY from degradation under
modified, or delayed, by additional polymer coatings, physiological digestive conditions. The decrease in
possessing different pH-release characteristics tailored activity of the non-encapsulated IgY following in
to the increasingly alkaline pH of the small intestine. vivo digestion is in agreement with the results of
Furthermore, it should be noted that the disintegration Yokoyama et al. (1993), who found that antibody
times of polymer films are often somewhat longer in activity decreased several fold after passage through
vivo than in vitro (Lehmann et al., 1999). the stomach of pigs.
To more accurately simulate gastrointestinal con- A greater amount of non-encapsulated IgY
ditions, the encapsulated IgY was first exposed to appeared to be present in the distal portion of the
SGF, before incubation with pancreatic enzymes in gastrointestinal tract, which may be due to the
the SIF. The non-encapsulated IgY was, however, sustained release characteristics of the microencapsu-
exposed only to the SIF, as it would have been lated sample. However, despite the larger quantities of
degraded completely in the SGF, making it impossible non-encapsulated IgY detected in the intestines, and
to assess the effects of the pancreatic enzymes. As the in contrast to the observations for the microencapsu-
polymer coating disintegrated in the SIF, the IgY was lated antibodies, its activity was disproportionately
brought into direct contact with the pancreatic lower, indicating that it had lost activity upon passage
enzymes. As expected, the simulated intestinal con- through the stomach.
208 J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209

The results demonstrate that the microencapsula- Eggum, B.O., Boisen, S., 1991. In vitro techniques of measuring
tion of IgY with an a pH-sensitive methacrylic acid digestion. In: Verstegen, M.W.A., Huisman, J., den Hartog, L.A.
(Eds.), Digestive Physiology in Pigs, Proceedings of the Vth
copolymer (EudragitR L100-55) can successfully International Symposium on Digestive Physiology in Pigs, EAAP
protect the antibodies from gastric inactivation, and Publication. vol. 54. Pudoc, Wageningen, Netherlands, p. 213.
may provide an effective means of controlling HRV Fallingborg, J., Christensen, L.A., Ingeman-Nielsen, M., Jacobsen,
infection, as well as having significant implications B.A., Abildgaard, K., Rasmussen, H.H., Rasmussen, S.N.,
1990. Measurement of gastrointestinal pH and regional transit
for passive immunization techniques in general, for
times in normal children. J. Pediatr. Gastroenterol. Nutr. 11, 211.
the widespread prevention of enteric pathogens. Hatta, H., Mabe, K., Kim, M., Yamamoto, T., Gutierrez, M.A.,
Miyazaki, T., 1993. Oral passive immunization effect of anti-
human rotavirus IgY and its behavior against proteolytic
enzymes. Biosci. Biotechnol. Biochem. 57, 1077.
Acknowledgements Hilpert, H., Brussow, H., Mietens, C., Sidoti, J., Lerner, L.,
Werchau, H., 1987. Use of bovine milk concentrate containing
This work was supported by the Natural Sciences antibody to rotavirus to treat rotavirus gastroenteritis in infants.
and Engineering Research Council of Canada J. Infect. Dis. 156, 158.
(NSERC), the Ontario Egg Producers Marketing Jensenius, J.C., Andersen, I., Hau, J., Crone, M., Koch, C., 1981.
Eggs: conveniently packaged antibodies. methods for purifica-
Board, Agriculture and Agri-Food Canada, and the tion of yolk IgG. J. Immunol. Methods 46, 63.
Ontario Ministry of Agriculture and Food (OMAF). Kapikian, A.Z., Chanock, R.M., 1996. Rotaviruses. In: Fields, B.N.,
We would like to thank Dr. Y. Takada, Snow Brand Knipe, D.N., Howley, P.M., Chanock, R.M., Melnick, J.L.,
Milk, Tokyo, Japan, for his technical support with the Monath, T.P., Rolzman, B., Strauss, S.E. (Eds.), Fields Virology
IgY granulation, and the staff at the Central Animal 3rd ed. Raven Press, New York, p. 1657.
Kararli, T.T., 1995. Comparison of the gastrointestinal anatomy,
Facility, Animal Care Services, University of Guelph, physiology, and biochemistry of humans and commonly used
for their assistance with the in vivo experiments. laboratory animals. Biopharm. Drug Dispos. 16, 351.
Kelly, C.P., Chetham, S., Keates, S., Bostwick, E.F., Roush, A.M.,
Castagliuolo, I., LaMont, J.T., Pothoulakis, C., 1997. Survival of
anti-Clostridium difficile bovine immunoglobulin concentrate in
References the human gastrointestinal tract. Antimicrob. Agents Chemother.
41, 236.
Akita, E.M., Nakai, S., 1992. Immunoglobulins from egg yolk: Kovacs-Nolan, J., Mine, Y., 2004. Avian egg antibodies: basic and
isolation and purification. J. Food Sci. 57, 629. potential applications. Avian Poult. Biol. Rev. 15, 25.
Akita, E.M., Nakai, S., 2000. Preparation of enteric-coated gelatin Kovacs-Nolan, J., Sasaki, E., Yoo, D., Mine, Y., 2001. Cloning and
capsules of IgY with cellulose acetate phthalate. In: Sim, J.S., expression of human rotavirus spike protein, VP8*, in
Nakai, S., Guenter, W. (Eds.), Egg Nutrition and Biotechnology. Escherichia coli. Biochem. Biophys. Res. Commun. 282, 1183.
CAB International, New York, p. 301. Laemmli, U.K., 1970. Cleavage of structural proteins during the
Bogstedt, A.K., Hammarstrfm, L., Robertson, A.-K., 1997. assembly of the head of the bacteriophage T4. Nature 227, 680.
Survival of immunoglobulins from different species through Lehmann, K., Agmus, M., Bfssler, H., Dreher, D., Liddiard, C.,
the gastrointestinal tract in healthy adult volunteers: implications Petereit, H.-U., Rothgang, G., Weisbrod, W., Beckert, T., 1999.
for human therapy. Antimicrob. Agents Chemother. 41, 2320. Practical Course in Film Coating of Pharmaceutical Dosage
Carlander, D., Kollberg, H., Wej3ker, P.-E., Larsson, A., 2000. Forms with EUDRAGITR. Rfhm, Pharma Polymers, Darms-
Peroral immunotherapy with yolk antibodies for the prevention tadt, Germany.
and treatment of enteric infections. Immunol. Res. 21, 1. Madsen, J.L., 1994. Gastrointestinal transit measurements, a
Castro, G.A., 1981. Digestion and absorption of specific nutrients. scintographic method. Dan. Med. Bull. 41, 398.
In: Johnson, E.R. (Eds.), Gastrointestinal Physiology, 2nd ed. Miller, E.R., Ullrey, D.E., 1987. The pig as a model for human
The C.V. Mosby, Toronto, p. 123. nutrition. Annu. Rev. Nutr. 7, 361.
Clark, H.F., Glass, R.I., Offit, P.A., 1999. Rotavirus vaccines. Mouecoucou, J., Villaume, C., Sanchez, C., Mejean, L., 2004. Beta-
In: Plotkin, S.A., Orenstein, W.A. (Eds.), Vaccines, 3rd ed. lactoglobulin/polysaccharide interactions during in vitro gastric
W.B. Saunders, Philadelphia, p. 987. and pancreatic hydrolysis assessed in dialysis bags of different
Davis, S.S., 1989a. Assessment of gastrointestinal transit and drug molecular weight cut-offs. Biochim. Biophys. Acta 1670, 105.
absorption. In: Prescott, L.F., Nimmo, W.S. (Eds.), Novel Drug Pacyna, J., Siwek, K., Terry, S.J., Roberton, E.S., Johnson, R.B.,
Delivery. John Wiley & Sons, Toronto, p. 89. Davidson, G.P., 2001. Survival of rotavirus antibody activity
Davis, S.S., 1989b. Small intestinal transit. In: Hardy, J.G., Davis, derived from bovine colostrums after passage through the
S.S., Wilson, C.G. (Eds.), Drug Delivery to the Gastrointestinal human gastrointestinal tract. J. Pediatr. Gastroenterol. Nutr.
Tract. John Wiley & Sons, Toronto, p. 49. 32, 162.
J. Kovacs-Nolan, Y. Mine / Journal of Immunological Methods 296 (2005) 199–209 209

Reilly, R.M., Domingo, R., Sandhu, J., 1997. Oral delivery of Sim, J.S., Sunwoo, H.H., Lee, E.N., 2000. Ovoglobulin IgY. In:
antibodies—future pharmacokinetic trends. Clin. Pharmacokinet. Naidu, A.S. Eds., Natural Food Antimicrobial systems. CRC
4, 313. Press, New York, p. 227.
Roos, N., Mahe, S., Benamouzig, R., Sick, H., Rautureau, J., Tome, United States Pharmacopeial Convention Council of Experts,
D., 1995. 15N-labeled immunoglobulins from bovine colostrums 2004a. Simulated gastric fluid, TS. In: Board of Trustees
are partially resistant to digestion in human intestine. J. Nutr. (Eds.), The United States Pharmacopeia 27, The National
125, 1238. Formulary, vol. 22. United States Pharmacopeial Convention,
Rowan, A.M., Moughan, P.J., Wilson, M.N., Maher, K., Tasman- Rockville, MD, p. 2728.
Jones, C., 1994. Comparison of the ileal and faecal digestibility of United States Pharmacopeial Convention Council of Experts,
dietary amino acids in adult humans and evaluation of the pig as a 2004b. Simulated intestinal fluid, TS. In: Board of Trustees
model animal for digestion studies in man. Br. J. Nutr. 71, 29. (Eds.), The United States pharmacopeia 27, The National
Shimizu, M., Nakane, Y., 1995. Encapsulation of biologically active Formulary, vol. 22. United States Pharmacopeial Convention,
proteins in a multiple emulsion. Biosci. Biotechnol. Biochem. Rockville, MD, p. 2728.
59, 492. Warr, G.W., Magor, K.E., Higgins, D.A., 1995. IgY: clues to the
Shimizu, M., Nagashima, H., Sano, K., Hashimoto, K., Ozeki, M., origins of modern antibodies. Immunol. Today 16, 392.
Tsuda, K., Hatta, H., 1992. Molecular stability of chicken and Yokoyama, H., Peralta, R.C., Sendo, S., Ikemori, Y., Kodama, Y.,
rabbit immunoglobulin G. Biosci. Biotechnol. Biochem. 56, 270. 1993. Detection of passage and absorption of chicken egg yolk
Shimizu, M., Nagashima, H., Hashimoto, K., 1993a. Comparative immunoglobulins in the gastrointestinal tract of pigs by use of
studies on molecular stability of immunoglobulin G from enzyme-linked immunosorbent assay and fluorescent antibody
different species. Comp. Biochem. Physiol. 106B, 255. testing. Am. J. Vet. Res. 54, 867.
Shimizu, M., Miwa, Y., Hashimoto, K., Goto, A., 1993b.
Encapsulation of chicken egg yolk immunoglobulin G (IgY)
by liposomes. Biosci. Biotechnol. Biochem. 57, 1445.

You might also like