You are on page 1of 5

European Journal of Pharmaceutical Sciences 44 (2011) 422–426

Contents lists available at SciVerse ScienceDirect

European Journal of Pharmaceutical Sciences


journal homepage: www.elsevier.com/locate/ejps

Chitosan nanoparticles enhance the plasma exposure of ( )-epigallocatechin


gallate in mice through an enhancement in intestinal stability
Admire Dube, Joseph A. Nicolazzo, Ian Larson ⇑
Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, Victoria 3052, Australia

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

Article history: The green tea catechin ( )-epigallocatechin gallate (EGCG) has attracted significant research interest due
Received 31 May 2011 to its beneficial therapeutic effects, which include anti-oxidant, neuro-protective and anti-cancer effects.
Received in revised form 24 August 2011 However, the therapeutic potential of EGCG following oral consumption is limited by its poor absorption.
Accepted 5 September 2011
To address this issue, EGCG has been encapsulated in chitosan-tripolyphosphate nanoparticles (CS NPs)
Available online 8 September 2011
and the oral absorption of EGCG evaluated in Swiss Outbred mice. Administration of the CS NPs enhanced
the plasma exposure of total EGCG by a factor of 1.5 relative to an EGCG solution, with plasma AUC(0 5 h)
Keywords:
values of 116.4 ± 4.1 and 179.3 ± 10.8 nM.h (mean ± s.d., n = 3 5) for the EGCG solution and CS NPs,
( )-Epigallocatechin gallate (EGCG)
Oral absorption
respectively. Associated with the increased plasma exposure of EGCG was an enhancement in concentra-
Plasma exposure tions of EGCG in the stomach and jejunum of mice following CS NP administration. A 2.3-fold increase in
Chitosan-tripolyphosphate nanoparticles the apparent exposure of EGCG to the jejunum (AUCj) was observed following CS NP encapsulation, with
(CS NPs) AUCj(0 5 h) values of 5.3 ± 1.1 and 12.3 ± 1.5 lM.h (mean ± s.d., n = 3 5) for the EGCG solution and CS
Intestinal stability NPs, respectively. The enhanced exposure of EGCG to the jejunum was likely responsible for the increased
plasma concentrations of EGCG. The findings from this study suggest that CS NPs may be a useful
approach for enhancing oral delivery, and therapeutic application, of EGCG in a number of disease
conditions.
Ó 2011 Published by Elsevier B.V.

1. Introduction However, following oral administration of EGCG to rodents at rel-


evant doses, peak plasma concentrations in the nM range are ob-
( )-Epigallocatechin gallate (EGCG) (Fig. 1), a flavonoid belong- tained (Chen et al., 1997; Lin et al., 2007; Dube et al., 2011), and
ing to the catechin class, is found in various foods and beverages, with even higher doses (equivalent to ingestion of multiple cups
particularly in green tea, where it is present in high amounts of tea), plasma concentrations of EGCG are at most 1 lM (Balentine
(Wang et al., 2000; Hackman et al., 2008). Recently, EGCG has et al., 1997; Raneva et al., 2005; Lambert et al., 2006a). The low oral
attracted research interest as several in vitro and animal studies absorption of EGCG, and that of catechins in general, has mainly
have demonstrated that it possesses favourable therapeutic effects, been attributed to poor stability within the gastro-intestinal tract
which include anti-oxidant, anti-viral, anti-inflammatory, cardio- (GIT) and poor permeability across the intestine (Zhu et al., 2000;
protective, neuro-protective and anti-cancer effects (Lambert and Cai et al., 2002; Chow et al., 2005; Lambert et al., 2006b; Huo
Yang, 2003; Nagle et al., 2006; Zaveri, 2006; Kuzuhara et al., et al., 2008; Wang et al., 2009). Catechins are unstable in gastro-
2008; Ho et al., 2009; Luo et al., 2010). intestinal solutions (Zhu et al., 1997; Huo et al., 2008), and as we
However, the therapeutic potential of EGCG is limited by its demonstrated recently, approximately 80% of EGCG degrades in
poor systemic absorption following oral consumption (Chen simulated intestinal fluid pH 7.4 (37 °C) over a 1 h period (Dube
et al., 1997; Lambert et al., 2003; Williamson, 2005; Lin et al., et al., 2010a). In addition to inherent instability, the apparent per-
2007; Huo et al., 2008). Studies indicate that, at most, 5% of an meability co-efficient (Papp) of EGCG across Caco-2 cells has been
EGCG dose is absorbed into the systemic circulation of rats follow- reported to be 0.83  10 7 cm/s (Zhang et al., 2004), suggesting
ing oral administration (Chen et al., 1997; Zhu et al., 2000; Lin very low permeability, as the Papp for the poorly-permeable marker
et al., 2007). It has been estimated that in order for EGCG to induce mannitol is at least 6.8  10 6 cm/s (Polentarutti et al., 1999; Ste-
relevant therapeutic effects, a concentration of at least 10 lM is phens et al., 2002; Yang et al., 2003).
required (Scalbert and Williamson, 2000; Yang et al., 2008). In order to fully maximise the therapeutic utility of EGCG, there
is a need to enhance its oral absorption (Huo et al., 2008; Schee-
⇑ Corresponding author. Tel.: +61 3 99039570; fax: +61 3 99039583. pens et al., 2009). Previous strategies to enhance the oral absorp-
E-mail address: ian.larson@monash.edu (I. Larson). tion of EGCG have included the formulation of a peracetate ester

0928-0987/$ - see front matter Ó 2011 Published by Elsevier B.V.


doi:10.1016/j.ejps.2011.09.004
A. Dube et al. / European Journal of Pharmaceutical Sciences 44 (2011) 422–426 423

De-ionised water was obtained from a MilliporeÒ filtration system


(Billerica, MA).

2.2. Encapsulation of EGCG in CS NPs

CS NPs encapsulating EGCG were prepared as previously de-


scribed (Dube et al., 2010b). Briefly, NPs were prepared by the
drop-wise addition of a tripolyphosphate (TPP) solution (0.1%, w/
v; pH 3) to a CS solution (0.1% w/v in 0.175% v/v acetic acid; pH
3) containing EGCG (0.05% w/v), under magnetic stirring for
10 min (700 rpm) at ambient temperature. NPs were collected by
centrifugation at 40,000g for 30 min and freeze-dried (Virtis
AdVantage™, USA) at 80 °C for 16 h. The CS NPs contained
3.7 ± 0.1 lg/mg of EGCG with an average size and zeta potential
of 440 ± 37 nm and 25 ± 1 mV, respectively, determined in Krebs
Fig. 1. Chemical structure of EGCG. Bicarbonate-buffered Ringers solution pH 6.0 (Dube et al., 2010b).

2.3. Dosing of mice and sample collection


pro-drug of EGCG, which is more stable than EGCG in solution
(Lam et al., 2004). Oral administration of peracetylated EGCG to
All animal experiments were approved by the Monash Institute
mice resulted in a 2.4-fold increase in the plasma exposure of
of Pharmaceutical Sciences Animal Ethics Committee and were
EGCG, relative to an EGCG solution (Lambert et al., 2006b). Another
performed in accordance with the Australian National Health and
approach has been the co-administration of EGCG with the com-
Medical Research Council guidelines for the care and use of ani-
pounds piperine or genistein. Piperine and genistein are able to in-
mals for scientific purposes. Male Swiss Outbred mice (25–30 g)
hibit the biotransformation and the intestinal efflux of EGCG,
were obtained from Monash Animal Services and were kept in
respectively (Lambert et al., 2004, 2008). With the use of these
air-conditioned quarters with a room temperature of 20 ± 2 °C, rel-
compounds, the plasma exposure of EGCG could be increased up
ative humidity of 50 ± 10% and an alternating 12 h light: dark cycle.
to 1.5-fold (Lambert et al., 2004, 2008). However, the use of pro-
Mice were fasted for 16 h prior to administration of the EGCG for-
drugs or co-administration of metabolism or efflux inhibitors
mulations. To assess the plasma exposure of EGCG from solution, a
may not be the most appropriate approaches due to possible tox-
volume of an EGCG solution (0.11 mg/ml in 0.1 M HCl) was admin-
icity arising from these compounds, or the impact of these agents
istered to mice by oral gavage equating to an EGCG dose of
on the oral absorption of toxins normally denied access by these
0.76 mg/kg (Dube et al., 2011). To assess the plasma exposure of
metabolizing enzymes or efflux transporters.
EGCG from the CS NPs, a suspension of the NPs (35 mg/ml in
Our approach to overcome the issue of poor oral bioavailability
0.1 M HCl) was prepared. The NPs were suspended in 0.1 M HCl
has been to encapsulate EGCG in chitosan nanoparticle (CS NP)
through sonication for 30 s and immediately thereafter, a volume
delivery systems. Previously, we reported that encapsulation in
of the suspension administered by oral gavage to mice to give an
CS NPs increases the stability of EGCG in simulated GIT fluids,
equivalent dose of 0.76 mg/kg EGCG. At pre-determined time
and leads to enhanced absorption of EGCG in an in vitro mouse
intervals post-dose (30, 60, 90, 150, 210, 300 and 360 min), mice
intestinal permeability model (Dube et al., 2010a,b). Our investiga-
(n = 3 5 at each time point) were anaesthetized with an intra-
tions suggested that the enhanced absorption of EGCG was exerted
peritoneal dose of ketamine (133 mg/kg) and xylazine (10 mg/
by enhanced stability imparted by the NPs on EGCG, leading to in-
kg), and blood collected by cardiac puncture into polyethylene
creased flux of EGCG across the intestine (Dube et al., 2010b). How-
tubes containing 10 ll of the anticoagulant heparin. Blood was
ever, the impact of the CS NPs on the oral absorption of EGCG
centrifuged at 16 000g for 5 min and 100 ll aliquots of plasma
in vivo remains unknown. Therefore, the aim of the present study
(n = 2) collected into polyethylene tubes containing 20 ll of
was to determine the impact of encapsulation of EGCG in CS NPs
20 mM AA and 13 mM TCEP stabilizing solution, which we have
on the plasma exposure of EGCG following administration to mice.
previously reported as an effective stabilizer of EGCG in aqueous
In addition, this study investigates whether any enhancement in
and plasma samples (Dube et al., 2010a,b). Plasma samples were
plasma exposure induced by the NPs is related to increased stabil-
snap frozen using carbon-dioxide dry ice and stored at 20 °C until
ity of EGCG in the GIT, as has been suggested from in vitro studies.
HPLC analysis. The rationale for collecting two aliquots of plasma
for each sample was to allow for determination of both un-conju-
2. Materials and methods gated EGCG (i.e. non-metabolized EGCG) and total EGCG (i.e. the
sum of non-metabolized, and glucuronidated and sulfated EGCG).
2.1. Chemicals On the day of the assay, EGCG was extracted from the plasma sam-
ples and quantified as described previously (Dube et al., 2011).
EGCG (95% purity), ascorbic acid (AA), tris(2-carboxyethyl) Briefly, to determine total EGCG, 5 ll of b-glucuronidase
phosphine (TCEP) and disodium-ethylenediaminetetraacetic acid (500,000 U/ml) and 5 ll of sulfatase (4000 U/ml) enzymes were
(Na2EDTA) were obtained from Sigma–Aldrich Co (St Louis, MO). added to the plasma sample and incubated at 37 °C for 45 min.
The enzymes b-D-glucuronidase (G-7896, EC 3.2.1.31, from Esche- To determine un-conjugated EGCG, 10 ll of K2HPO4 buffer were
richia coli with 9  106 U/g solid) and sulfatase (S-9754, EC 3.1.6.1, added in place of the enzymes, and the sample also incubated for
from Abalone entrails with 2.3  105 U/g solid) were also obtained 45 min. After incubation, EGCG was extracted from the samples
from Sigma Aldrich Co (St Louis, MO). Stock solutions of these using ethyl acetate. The ethyl acetate layer containing EGCG was
enzymes were prepared in 50 mM K2HPO4 buffer and stored at separated from the sample, evaporated, and the residue reconsti-
20 °C until required. Acetic acid (glacial) was obtained from Ajax tuted with 100 ll of 15% v/v aqueous acetonitrile solution. A
Finechem (Taren Point, Australia) and HPLC grade acetonitrile was 50 ll aliquot of this solution was injected onto the HPLC column
purchased from Merck (Darmstadt, Germany). All other chemicals and EGCG was quantified using a validated HPLC/UV method de-
and solvents were of analytical grade and were used as received. scribed previously (Dube et al., 2011).
424 A. Dube et al. / European Journal of Pharmaceutical Sciences 44 (2011) 422–426

Table 1
Plasma pharmacokinetic parameters of EGCG (un-conjugated and total) following oral administration of an EGCG solution and CS NPs to Swiss Outbred mice at a dose of 0.76 mg/
kg.

Parameter EGCG solution CS NP


Un-conjugated Total Un-conjugated Total
AUC(0 5 h) (nM.h) 114.3 ± 4.1⁄ 116.4 ± 4.1⁄⁄ 166.0 ± 12.0⁄ 179.3 ± 10.8⁄⁄
Cmax (nM) 31.5 ± 3.3 34.3 ± 2.0 35.1 ± 9.2 37.8 ± 6.8
Tmax (h) 1.5 1.5 1.5 1.5

Data shown are the mean ± s.d. from 3–5 animals. Statistically significant differences in the plasma exposure between the EGCG solution and the CS NPs are indicated by ⁄ for
unconjugated EGCG and ⁄⁄ for total EGCG, respectively (z > 1.96).

2.3.1. Assessment of the impact of the CS NPs on the stability of EGCG significant differences (p > 0.05) between the concentrations of
in the GIT of the mice un-conjugated and total EGCG in the plasma samples were ob-
To evaluate whether encapsulation in CS NPs was associated served, suggesting that minimal metabolism of EGCG occurred in
with enhanced stability of EGCG within the GIT of mice, the con- mice, as has previously been reported (Meng et al., 2002). The en-
centration of EGCG in the stomach and jejunum of dosed mice hanced plasma exposure of EGCG through the use of CS NPs may
was determined. Following administration of the EGCG solution provide some benefit for the treatment of systemic disorders
or the CS NPs (as described in Section 2.3.1), samples were ob- where EGCG is suggested to exert therapeutic activity, albeit fur-
tained from mice sacrificed at the 90, 150 and 300 min time points ther studies may be required to obtain plasma concentrations clo-
(n = 3 5 at each time point). To obtain gastric samples, the stom- ser to therapeutically-active concentrations.
ach was isolated and an incision made along the midline, exposing Fig. 2 shows the plasma concentration versus time profile of
the inner contents of the stomach. Using a flat metal spatula, the EGCG obtained following administration of the EGCG solution
contents of the stomach were thoroughly scraped and collected and the CS NPs to mice. It was observed that administration of
into polyethylene tubes containing 1 ml of 20 mM AA and 13 mM the EGCG solution resulted in plasma concentrations which peaked
TCEP stabilizing solution. To obtain jejunal samples, the jejunum at approximately 1.5 h and after this period declined to signifi-
(i.e. the segment beginning 5 cm distal to the ligament of Treitz cantly lower levels (i.e. p < 0.05 for plasma concentrations between
and extending distally for 10 cm (Stephens et al., 2002)), was re- 1.5 and 5 h). However, following administration of the CS NPs,
moved and using a syringe, flushed thoroughly with 20 mM AA plasma concentrations of EGCG peaked at approximately 1.5 h
and 13 mM TCEP stabilizing solution into a polyethylene tube. and were sustained up to 5 h (i.e. p > 0.05 for plasma concentra-
Samples were vortexed for 1 min before being filtered through a tions between 1.5 and 5 h). At the 1.5 h time point, plasma concen-
0.1 lm filter and a 50 ll aliquot of the filtrate kept for assay. EGCG trations of EGCG were not significantly different between
that was free in solution, i.e. not encapsulated in the NPs, was as- administration of the EGCG solution and the CS NPs (Table 1). This
sayed according the validated HPLC/UV method (Dube et al., suggested that the CS NPs may not have enhanced the plasma
2010a). exposure of EGCG through increasing the permeability of EGCG
but rather through providing sustained concentrations of EGCG
in the GIT for subsequent availability in the plasma. Such a ratio-
2.4. Data analysis
nale was inferred as it was anticipated that an increase in intestinal
permeability would lead to an increase in the peak plasma concen-
Statistical differences between the various experimental treat-
trations of EGCG and/or a decrease in the time to reach peak plas-
ments were determined using the independent samples student’s
ma concentrations (Damgé et al., 2007; Sarmento et al., 2007). As
t test or the analysis of variance (ANOVA) test using SPSS Statistics
the CS NPs had a mean size of 440 ± 37 nm, it was unlikely that
v17.0 software (Chicago, IL). Values of p < 0.05 indicated statisti-
the particles could have enhanced the plasma exposure by being
cally significant differences. The plasma exposure of EGCG follow-
ing administration of the EGCG solution and the CS NPs was
determined through calculation of the area under the plasma con-
centration versus time curve from time zero to 5 h post-dose
(AUC0 5 h) using Bailer’s method (Bailer, 1988). Statistical differ-
ences between the AUC of un-conjugated or total EGCG were deter-
mined using the statistical z test, where z values greater than 1.96
indicated statistically significant differences (Bailer, 1988; Yuan,
1993; Nedelman et al., 1995). The peak plasma concentration
(Cmax) and time of peak concentration (Tmax) were obtained di-
rectly from the plasma concentration vs time profiles.

2.5. Results and discussion

2.5.1. Plasma exposure of EGCG and impact of CS NP encapsulation


The plasma exposure of EGCG in mice was determined follow-
ing oral administration of the EGCG solution and the CS NPs and
the associated pharmacokinetic parameters for EGCG are listed in
Table 1.
Administration of the CS NPs resulted in a significantly en- Fig. 2. Plasma concentrations of total EGCG after oral administration of an EGCG
solution (-d-) and CS NP formulation (-s-) to Swiss Outbred mice (0.76 mg/kg).
hanced plasma exposure (AUC(0 5 h)) of both un-conjugated and Data shown are the mean ± s.e.m. from 3–5 animals. For clarity, profiles of total
total EGCG, relative to the EGCG solution (z > 1.96), with the CS EGCG only are shown as there was no significant difference (p < 0.05) between the
NPs leading to a 1.5-fold higher plasma exposure. No statistically plasma concentrations of un-conjugated and total EGCG.
A. Dube et al. / European Journal of Pharmaceutical Sciences 44 (2011) 422–426 425

Fig. 3. Concentrations of EGCG measured in samples obtained from the stomach and jejunum of Swiss Outbred mice following oral administration of the EGCG solution
( ) and the CS NPs formulation ( ). In both treatments, the dose of EGCG administered to mice was 0.76 mg/kg. Data are the mean ± s.e.m. from 3–5 animals.

Indicates statistical differences in concentration between administration of the EGCG solution and the CS NPs.

taken up across the intestinal wall through the process of transcy- completely released EGCG (un-published data) and hence, the ef-
tosis. This is because only particles with a mean size under 100 nm fect of the CS NPs on the stabilization of EGCG in the jejunum
and particularly those approaching 50 nm, have the potential to be dissipates.
taken up by intestinal cells (Florence et al., 1995; Florence and It is possible that delayed gastric emptying following adminis-
Hussain, 2001; Barratt, 2003). In addition, we reported that these tration of the CS NPs could have contributed to the enhanced con-
CS NPs were unable to enhance the paracellular or passive centrations of EGCG. To determine the impact of gastric emptying,
transcellular transport of compounds across the intestinal tissue an assessment of the relative concentrations of EGCG in the stom-
of mice (Dube et al., 2010b), and therefore it was unlikely that ach samples to those in the jejunum samples, after administration
the enhanced plasma exposure of EGCG with CS NP was due to of the EGCG solution or the CS NPs was conducted (Table 2).
increased intestinal permeability. Rather, the enhancement in the No significant difference (p > 0.05) in the stomach: jejunum
plasma exposure of EGCG was likely to be due to other effects of EGCG concentration ratios at each time point following adminis-
the CS NPs, such as an enhancement in the stability of EGCG within tration of the EGCG solution or the CS NPs was observed. It was ex-
the GIT leading to increased flux across the intestine, as we have pected that had delayed gastric emptying occurred, the ratio of the
previously reported from in vitro studies (Dube et al., 2010b). To stomach: jejunum EGCG concentrations following administration
determine whether the CS NPs also enhanced the stability of EGCG of the CS NPs would have been significantly higher than observed
in vivo, the concentrations of EGCG in the GIT of the mice were also following administration of the EGCG solution, due to the accumu-
determined. lation of EGCG in the stomach. Therefore, the results suggested that
delayed gastric emptying could be excluded as a factor enhancing
2.5.2. Assessment of the impact of CS NPs on the stability of EGCG in the gastro-intestinal concentrations of EGCG.
the stomach and jejunum of mice The jejunum has been reported as the main site of absorption
Concentrations of EGCG in samples collected from the stomach for flavonoid compounds (Matuschek et al., 2006). Consequently,
and jejunum of mice following oral administration of the EGCG the ‘apparent’ exposure of the jejunal contents to EGCG was deter-
solution and the CS NPs were determined. Fig. 3 shows the concen- mined, through calculation of the area under the jejunal concentra-
trations of EGCG at 1.5, 2.5 and 5 h post dose. It should be noted tion vs time profiles (AUCj(0 5 h)). AUCj(0 5 h) was calculated
that the EGCG concentrations measured were reflective of concen- using Bailers approach as described in Section 2.4. AUCj(0 5 h)
trations in the sample of AA/TCEP solution, and were not the actual was 5.3 ± 1.1 and 12.3 ± 1.5 lM.h (mean ± s.d., n = 3–5) following
concentrations of EGCG present in the gut segments. However, gi- administration of the EGCG solution and the CS NPs, respectively,
ven that samples from both the EGCG solution and CS NP-treated indicating that administration of the CS NPs had resulted in a
mice were treated in an identical manner, any concentration differ- 2.3-fold increase in the ‘apparent’ exposure of the jejunum to EGCG
ences observed were reflective of the in vivo situation. over the 5 h period. The enhanced exposure of the jejunum to
It was observed that at 1.5 and 2.5 h following administration of EGCG was likely responsible for the enhancement in the plasma
the CS NPs, EGCG gastric concentrations were significantly higher exposure of EGCG, and was in agreement with the findings from
than those detected following administration of the EGCG solution previous in vitro absorption studies, where significantly higher
(p < 0.05), whereas after 5 h, the concentrations were similar
(p > 0.05). In the jejunum, EGCG concentrations at the 2.5 h time Table 2
point were significantly higher following administration of the CS The ratios of the concentrations of EGCG measured in the stomach samples to those
NPs compared to the EGCG solution (p < 0.05), whereas at 1.5 measured in the jejunum samples at each of the sampling time points, following oral
administration of the EGCG solution or the CS NPs to Swiss Outbred mice. Data shown
and 5 h, the concentrations were similar (p > 0.05). These observa-
are mean ratios ± (s.d.) (i.e. x: 1), calculated from 3–5 animals.
tions suggested that encapsulation of EGCG in CS NPs enhanced its
stability in the GIT of mice (in vivo), as we have previously Time (min) EGCG solution CS NPs

observed in vitro (Dube et al., 2010a,b). In-vivo, the stabilization Mean ratio (stomach: jejunum)
occurs firstly in the stomach and later in the jejunum as the CS 90 2.0 ± 0.1 4.9 ± 2.9
150 1.2 ± 0.9 1.3 ± 0.7
NPs require time to move from the stomach into the small intes-
300 0.9 ± 0.1 0.9 ± 0.2
tine and to release EGCG. By 5 h, it is likely that the CS NPs have
426 A. Dube et al. / European Journal of Pharmaceutical Sciences 44 (2011) 422–426

concentrations of EGCG were present on the mucosal side of the Lambert, J.D., Yang, C.S., 2003. Mechanisms of Cancer Prevention by Tea
Constituents. Journal of Nutrition 133 (10), 3262S–3267S.
jejunum following encapsulation in CS NPs (Dube et al., 2010b).
Lambert, J.D., Lee, M.-J., et al., 2003. Epigallocatechin-3-gallate is absorbed but
The enhanced exposure of the jejunum to EGCG following admin- extensively glucuronidated following oral administration to mice. Journal of
istration of the CS NPs could be potentially beneficial for inflamma- Nutrition 133 (12), 4172–4177.
tory conditions of the intestine such as Crohn’s disease, where Lambert, J.D., Hong, J., et al., 2004. Piperine enhances the bioavailability of the tea
polyphenol ( )-epigallocatechin-3-gallate in mice. The Journal of Nutrition 134
prolonged exposure to high concentrations of antioxidants such (8), 1948–1952.
as EGCG is required (Chapple, 1997; Porath et al., 2005). In addi- Lambert, J.D., Lee, M.-J., et al., 2006a. Dose-dependent levels of Epigallocatechin-3-
tion, other locally and systemically-acting therapeutic compounds gallate in human colon cancer cells and mouse plasma and tissues. Drug
Metabolism and Disposition 34 (1), 8–11.
which exhibit poor stability could be encapsulated in CS NPs to Lambert, J.D., Sang, S., et al., 2006b. Peracetylation as a means of enhancing in-vitro
enhance their stability and consequently, their therapeutic effec- bioactivity and bioavailability of epigallocatechin-3-gallate. Drug Metabolism
tiveness either in the GIT or in the systemic circulation. and Disposition 34 (12), 2111–2116.
Lambert, J.D., Kwon, S.-J., et al., 2008. Effect of genistein on the bioavailability and
intestinal cancer chemopreventive activity of ( )-epigallocatechin-3-gallate.
Carcinogenesis 29 (10), 2019–2024.
3. Conclusion Lin, L.-C., Wang, M.-N., et al., 2007. Pharmacokinetics of ( )-epigallocatechin-3-
gallate in conscious and freely moving rats and its brain regional distribution.
Oral administration of EGCG encapsulated in CS NPs resulted in Journal of Agricultural and Food Chemistry 55 (4), 1517–1524.
Luo, T., Wang, J., et al., 2010. ( )-Epigallocatechin gallate sensitizes breast cancer
a significant enhancement in the plasma exposure of EGCG relative
cells to paclitaxel in a murine model of breast carcinoma. Breast Cancer
to an EGCG solution. The enhancement in oral absorption was Research 12 (1), R8.
attributed to an increased exposure of EGCG to the jejunum due Matuschek, M.C., Hendriks, W.H., et al., 2006. The jejunum is the main site of
absorption for anthocyanins in mice. The Journal of Nutritional Biochemistry 17
to the stability enhancing effect of the CS NPs. CS NPs may there-
(1), 31–36.
fore enable the future therapeutic application of EGCG in a number Meng, X., Sang, S., et al., 2002. Identification and characterization of methylated and
of disease conditions. ring-fission metabolites of tea catechins formed in humans, mice, and rats.
Chemical Research in Toxicology 15 (8), 1042–1050.
Nagle, D.G., Ferreira, D., et al., 2006. Epigallocatechin-3-gallate (EGCG): Chemical
References and biomedical perspectives. Phytochemistry 67 (17), 1849–1855.
Nedelman, J.R., Gibiansky, E., et al., 1995. Applying Bailer’s method for AUC
Bailer, A.J., 1988. Testing for the equality of area under the curves when using confidence intervals to sparse sampling. Pharmaceutical Research 12 (1), 124–
destructive measurement techniques. Journal of Pharmacokinetics and 128.
Pharmacodynamics 16 (3), 303–309. Polentarutti, B.I., Peterson, A.L., et al., 1999. Evaluation of viability of excised rat
Balentine, D.A., Wiseman, S.A., et al., 1997. The chemistry of tea flavonoids. Critical intestinal segments in the Ussing Chamber: Investigation of morphology,
Reviews in Food Science and Nutrition 37 (8), 693–704. electrical parameters, and permeability characteristics. Pharmaceutical
Barratt, G., 2003. Colloidal drug carriers: achievements and perspectives. Cellular Research 16 (3), 446–454.
and Molecular Life Sciences (CMLS) 60 (1), 21–37. Porath, D., Riegger, C., et al., 2005. Epigallocatechin-3-gallate impairs chemokine
Cai, Y., Anavy, N.D., et al., 2002. Contribution of presystemic hepatic extraction to production in human colon epithelial cell lines. Journal of Pharmacology and
the low oral bioavailability of green tea catechins in rats. Drug Metabolism and Experimental Therapeutics 315 (3), 1172–1180.
Disposition 30 (11), 1246–1249. Raneva, V.G., Shimizu, Y., et al., 2005. Antioxidant activity in plasma and tissue
Chapple, I.L.C., 1997. Reactive oxygen species and antioxidants in inflammatory distribution of ( )-epigallocatechin gallate after oral administration to rats.
diseases. Journal of Clinical Periodontology 24 (5), 287–296. Journal of Oleo Science 54 (5), 289–298.
Chen, L., Lee, M.-J., et al., 1997. Absorption, distribution, and elimination of tea Sarmento, B., Ribeiro, A., et al., 2007. Oral bioavailability of insulin contained in
polyphenols in rats. Drug Metabolism and Disposition 25 (9), 1045–1050. polysaccharide nanoparticles. Biomacromolecules 8 (10), 3054–3060.
Chow, H.-H.S., Hakim, I.A., et al., 2005. Effects of dosing condition on the oral Scalbert, A., Williamson, G., 2000. Dietary intake and bioavailability of polyphenols.
bioavailability of green tea catechins after single-dose administration of The Journl of Nutrition 130, 2073S–2085S.
polyphenon E in healthy individuals. Clinical Cancer Research 11 (12), 4627– Scheepens, A., Tan, K., et al., 2009. Improving the oral bioavailability of beneficial
4633. polyphenols through designed synergies. Genes & Nutrition 5 (1), 75–87.
Damgé, C., Maincent, P., et al., 2007. Oral delivery of insulin associated to polymeric Stephens, R.H., Tanianis-Hughes, J., et al., 2002. Region-dependent modulation of
nanoparticles in diabetic rats. Journal of Controlled Release 117 (2), 163–170. intestinal permeability by drug efflux transporters: In-vitro studies in mdr1a( /
Dube, A., Ng, K., et al., 2010a. Effective use of reducing agents and nanoparticle ) mouse intestine. The Journal of Pharmacology and Experimental
encapsulation in stabilizing catechins in alkaline solution. Food Chemistry 122 Therapeutics 303 (3), 1095–1101.
(3), 662–667. Wang, H., Provan, G.J., et al., 2000. Tea flavonoids: their functions, utilisation and
Dube, A., Nicolazzo, J.A., et al., 2010b. Chitosan nanoparticles enhance the intestinal analysis. Trends in Food Science & Technology 11 (4-5), 152–160.
absorption of the green tea catechins (+)-catechin and ( )-epigallocatechin Wang, X., Wang, Y.-W., et al. (2009). Enhancing stability and oral bioavailability of
gallate. European Journal of Pharmaceutical Sciences 41 (2), 219–225. polyphenols using nanoemulsions. Micro/Nanoencapsulation of Active Food
Dube, A., Nicolazzo, J.A., et al., 2011. Assessment of plasma concentrations of ( )- Ingredients. Washington, DC, American Chemical Society: 198–212.
epigallocatechin gallate in mice following administration of a dose reflecting Williamson, G.G., 2005. Bioavailability and bioefficacy of polyphenols in humans. II.
consumption of a standard green tea beverage. Food Chemistry 128 (1), 7–13. Review of 93 intervention studies. The American Journal of Clinical Nutrition 81
Florence, A.T., Hussain, N., 2001. Transcytosis of nanoparticle and dendrimer (1S), 243S.
delivery systems: evolving vistas. Advanced Drug Delivery Reviews 50 Yang, H., Finaly, R., et al., 2003. Alteration in epithelial permeability and ion
(Supplement 1), S69–S89. transport in a mouse model of total parenteral nutrition. Critical Care Medicine
Florence, A.T., Hillery, A.M., et al., 1995. Nanoparticles as carriers for oral peptide 31 (4), 1118–1125.
absorption: studies on particle uptake and fate. Journal of Controlled Release 36 Yang, C.S., Sang, S., et al., 2008. Bioavailability issues in studying the health effects of
(1-2), 39–46. plant polyphenolic compounds. Molecular Nutrition & Food Research 52 (S1),
Hackman, R., Polagruto, J., et al., 2008. Flavanols: digestion, absorption and S139–S151.
bioactivity. Phytochemistry Reviews 7 (1), 195–208. Yuan, J., 1993. Estimation of variance for AUC in animal studies. Journal of
Ho, H.-Y., Cheng, M.-L., et al., 2009. Antiviral Effect of Epigallocatechin Gallate on Pharmaceutical Sciences 82 (7), 761–763.
Enterovirus 71. Journal of Agricultural and Food Chemistry 57 (14), 6140– Zaveri, N.T., 2006. Green tea and its polyphenolic catechins: medicinal uses in
6147. cancer and noncancer applications. Life Sciences 78 (18), 2073–2080.
Huo, C., Wan, S., et al., 2008. The challenge of developing green tea polyphenols as Zhang, L., Zheng, Y., et al., 2004. Investigation of intestinal absorption and
therapeutic agents. Inflammopharmacology 16 (5), 248–252. disposition of green tea catechins by Caco-2 monolayer model. International
Kuzuhara, T., Suganuma, M., et al., 2008. Green tea catechin as a chemical chaperone Journal of Pharmaceutics 287 (1-2), 1–12.
in cancer prevention. Cancer Letters 261 (1), 12–20. Zhu, Q.Y., Zhang, A., et al., 1997. Stability of green tea catechins. Journal of
Lam, W.H., Kazi, A., et al., 2004. A potential prodrug for a green tea polyphenol Agricultural and Food Chemistry 45 (12), 4624–4628.
proteasome inhibitor: evaluation of the peracetate ester of ( )-epigallocatechin Zhu, M., Chen, Y., et al., 2000. Oral absorption and bioavailability of tea catechins.
gallate [( )-EGCG]. Bioorganic & Medicinal Chemistry 12 (21), 5587–5593. Planta Medica 66 (5), 444–447.

You might also like