You are on page 1of 7

International Journal of Pharmaceutics 436 (2012) 311–317

Contents lists available at SciVerse ScienceDirect

International Journal of Pharmaceutics


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

Biopharmaceutics classification and intestinal absorption study of apigenin


Jianjun Zhang a , Dapeng Liu a , Yanting Huang a , Yuan Gao b,∗ , Shuai Qian c,∗∗
a
School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
b
School of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing 210009, PR China
c
School of Pharmacy, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China

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

Article history: The aim of the study was to characterize the biopharmaceutics classification system (BCS) category of
Received 21 March 2012 apigenin (AP) using intrinsic dissolution rate (IDR) and rat intestinal permeability, and to investigate the
Received in revised form 30 May 2012 intestinal absorption mechanism of AP in rats. In the present investigation, equilibrium solubility and
Accepted 2 July 2012
intrinsic dissolution rate (IDR) of AP were estimated in phosphate buffers. Effective intestinal permeability
Available online 13 July 2012
(Peff ) of AP was determined using single-pass intestinal perfusion (SPIP) technique in four segments
(duodenum, jejunum, ileum and colon) of rat intestine at three concentrations (10, 50 and 100 ␮g/ml).
Keywords:
The aqueous solubility of AP in tested phosphate buffers was very poor with maximum solubility of
Apigenin
Biopharmaceutics classification system
2.16 ␮g/ml at pH 7.5. The IDR of AP was very low with a value of 0.006 mg/min/cm2 . The minimum and
Intrinsic dissolution rate maximum Peff s determined by SPIP were 0.198 × 10−4 and 0.713 × 10−4 cm/s at jejunum and duodenum
Permeability site, respectively. In addition, the concentration-dependent permeability behavior was observed in the
Single-pass intestinal perfusion duodenum and jejunum, which suggested that AP was transported by both passive and active carrier-
mediated saturable mechanism in these two intestinal segments. However, the observed concentration-
independent permeability behavior in ileum and colon indicated primarily passive transport mechanism
of absorption of AP in the last two intestinal segments. AP was classified as class II drug of the BCS due
to its low solubility and high intestinal permeability. AP could be well absorbed in the whole intestine
with the main absorption site at duodenum. The absorption of AP in four intestinal segments exhibited
different transport mechanisms.
© 2012 Elsevier B.V. All rights reserved.

1. Introduction been included in various guidance documents of regulatory impor-


tance (FDA, 2000; WHO, 2005).
Since the biopharmaceutics classification system (BCS) for cor- According to the FDA guidance for biowaiver request of
relating in vitro dissolution and in vivo bioavailability was proposed immediate-release solid oral dosage forms, a drug substance is con-
in 1990s based on drug dissolution and gastrointestinal perme- sidered as highly soluble when its highest dose strength could be
ability, which were two fundamental parameters controlling the dissolved in ≤250 ml of aqueous media over the pH range of 1.0–7.5
rate and extent of drug absorption, it has had an increasing impact (FDA, 2000). However, the choice of dose has a great impact on
on regulatory practice (Amidon et al., 1995). According to the BCS, the calculation of the dose/solubility ratio and, for some drugs, the
drug substances are classified as follows: high solubility–high per- classification may shift from ‘highly’ soluble to ‘not highly’ sol-
meability drugs (Class I), low solubility–high permeability drugs uble when using different dose in calculation (Lindenberg et al.,
(Class II), high solubility–low permeability drugs (Class III) and low 2004; Yu et al., 2004). In addition, solubility, as a static equilibrium
solubility–low permeability drugs (Class IV). Each class of the BCS parameter, cannot adequately describe the dynamic behavior of the
has its designated rate-limiting step and the reasonable strategies dissolution process especially in the physiological condition (Rinaki
for its modification that enable the formulator to select and opti- et al., 2003). Recently, dissolution-based classifications including
mize a dosage form for the specified drug (Cook et al., 2008; Ku, dissolution index (DI) (Papadopoulou et al., 2008) and intrinsic
2008). The BCS also provides a regulatory tool for the replacement dissolution ratio (IDR) (Yu, 2004; Zakeri-Milani, 2009) have been
of certain expensive and time-consuming bioequivalence studies developed for new molecular entities and marketed drugs, instead
by conducting accurate in vitro dissolution tests, and it has also of dose/solubility ratio based classification. Mean intestinal transit
time and mean time calculations for drug dissolution are included
in the dissolution parameter DI, which takes account the physiolog-
∗ Corresponding author. Tel.: +86 25 83379418; fax: +86 25 83379418. ical conditions (Papadopoulou et al., 2008). IDR is generally defined
∗∗ Corresponding author. Tel.: +852 51387572. as the dissolution rate of a pure drug substance under the condition
E-mail addresses: amicute@163.com (Y. Gao), sqian@cuhk.edu.hk (S. Qian). of constant surface area, agitation or stirring speed, pH and ionic

0378-5173/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.ijpharm.2012.07.002
312 J. Zhang et al. / International Journal of Pharmaceutics 436 (2012) 311–317

7.5 (pH value was adjusted by H3 PO4 (HCl for pH 1) or NaOH) (Wu
et al., 2005). In brief, an excess amount of AP was added into 10 ml
of buffer solution in a vial placed in a 37 ◦ C water bath. Following
equilibrium with shaking for 3 days, samples were filtered through
0.22 ␮m PTFE filter prior to analysis by the validated HPLC/UV
method. In each buffer, the solubilities of AP were analyzed in
triplicates.

Fig. 1. Chemical structure of AP.


2.3. Procedure of IDR measurement

An improved method for IDR measurement was employed


strength of the dissolution medium. The true IDR may be better to perform the study (Wood et al., 1965). Briefly, a quantity of
described as the rate of mass transfer from the solid surface to the 150 mg of AP was compressed with a compression force of 1.20 MPa
liquid phase. Since IDR is a rate phenomenon like permeability, it for 1 min to make non-disintegrating compacts using die and
might correlate better with in vivo drug dissolution rate than the punch (diameter: 8 mm). The surface area of the compacts was
equilibrium solubility (Yu et al., 2004). 0.5024 cm2 . Compacts were placed in a molten beeswax-mold in
To determining drug permeability in gastrointestinal tract, lots such a way that only one face could be in contact with dissolution
of in vivo, in vitro and in situ experimental models have been medium.
developed (Hillgren et al., 1995; Egan and Lauri, 2002; Refsgaard USP II dissolution apparatus (Sotax A7 Dissolution Apparatus:
et al., 2005; Varma and Panchagnula, 2005). Among these models, Sotax Ltd., London, UK) was applied in the in vitro dissolution
the single-pass intestinal perfusion (SPIP) method provides in situ study. Dissolution tests (six replicates) were performed in 900 ml of
experimental conditions simulating oral administration to investi- 0.05 mol/l phosphate buffer (pH = 6.8) at 37 ◦ C. The paddle rotating
gate the absorptive potential and mechanism of absorption of the speed was set at 100 rpm. Samples were collected through 0.22 ␮m
drug, where the innervations and clearance capabilities of the ani- syringe filters every 30 min for a period of 8 h. The obtained samples
mal remain intact. In addition, SPIP offers a simple and relevant were analyzed by the validated HPLC/UV method.
method of permeability assessment and correlates significantly
with the true absorption properties in human beings (Amidon et al., 2.4. Perfusion study using rat single-pass intestinal perfusion
1988; Logan, 2009). model
Apigenin (AP), 5,7-dihydroxy-2-(4-hydroxyphenyl)-4H-1-ben-
zopyran-4-one (Fig. 1), is one of the active ingredients found 2.4.1. Animals and surgical procedures
naturally in a variety of fruits and vegetables (Peterson and The study was approved by the Ethical Committee of China
Dwyer, 1998). It is recognized in traditional medicine and herbal Pharmaceutical University. Male Sprague-Dawley rats weighting
supplements for its various pharmacological activities, including between 230 and 270 g were obtained from the Laboratory Animal
anti-inflammatory effects (Funakoshi-Tago et al., 2011), free radi- Center, China Pharmaceutical University (Nanjing, China). Animals
cal scavenging effects (Horvathova et al., 2003), growth inhibitory were housed in standard cages on 12 h light–dark cycles, fed with
properties in several cancer lines including breast (Yin et al., 2001), standard animal chow and tap water daily. Prior to the experi-
colon (Wang et al., 2000), skin (Tong et al., 2007), myeloid leukemia ments, the animals were subjected to fasting for 12 h but allowed
cells (Takahashi et al., 1998) and pancreas (Ujiki et al., 2006). free access to water. All animals used in this study were handled
Moreover, recent clinical studies demonstrated that AP intake was in accordance with the guidelines of the Principles of Laboratory
associated with a suggestive decrease in woman’s risk of ovarian Animal Care (State Council, revised 1988).
cancer (Gates et al., 2007, 2009), which indicated AP could be a very SPIP studies were performed according to established meth-
promising drug candidate for future drug development. However, ods as previously described (Zakeri-Milani et al., 2009; Svensson
both of the basic physicochemical and biopharmaceutical proper- et al., 1999). In brief, the rats were anaesthetized by pentobar-
ties of AP were still limited from the published literatures. There is bital sodium (i.p., 50 mg/kg) and placed on a heated pad to keep
a need to classify the AP into BCS in order to facilitate the future for- body temperature. After making a 3–4 cm midline abdominal inci-
mulation development according to reasonable strategies of each sion, the small intestinal was exposed and then duodenal, jejunum,
BCS class drugs. In the present study, solubility, IDR property and ileum and colon were identified as follows: duodenum segment
permeability of AP were investigated to classify AP into a particular beginning from pylorus, jejunum segment beginning from 25 cm
BCS class, and the intestinal absorption mechanism of AP was also away from pylorus, while ileum segment the beginning at the site
studied. 20 cm upwards caecum, colon segment beginning from caecum
empennage. The intestinal segment of interest was located and
2. Materials and methods cannulated with silicon tubing connected to a peristaltic pump
(infusion pump, BT100-2G, Longer Pump, China). The outlet tubing
2.1. Materials was placed 10–12 cm aboral to the opening. The segment was then
rinsed with warm saline to remove intestinal contents and approx-
Apigenin (purity > 99.0% by HPLC) was provided by Nanjing imately 10 cm of the inlet tubing was placed inside the abdominal
Zelang Medical Technology Co., Ltd. (Nanjing, China). Tween 80 and cavity to achieve an inlet perfusion solution at 37 ◦ C. The whole
pentobarbital sodium were purchased from Guoyao Chemical Co., surgical area was covered with a piece of sterilized gauze wetted
Ltd. (Shanghai, China). HPLC grade water was produced by a Milli-Q with saline solution. The exposed intestinal segment was covered
water purification system (Millipore Co., Ltd., Bedford, USA). Other with Parafilm to decrease moisture loss.
chemicals were of HPLC or analytical grade. The experiment was initiated by rapidly filling the segment with
perfusion solution and the time was set zero with the immediate
2.2. Solubility determination start of the perfusion. The perfusion rate was set at 0.2 ml/min.
Care was taken to handle the small intestine gently and to mini-
The equilibrium solubilities of AP were determined in mize the surgery in order to maintain an intact blood supply. After
0.1 mol/ml phosphate buffers over pH value ranging from 1.0 to approximately 30 min, when steady-state was achieved, the outlet
J. Zhang et al. / International Journal of Pharmaceutics 436 (2012) 311–317 313

perfused samples were collected at 15 min intervals up to 120 min. Table 1


Solubility of apigenin in phosphate buffers (n = 3).
The perfusate was collected every 15 min into microtube and cen-
trifugated at 10000 rpm for 10 min. Samples were stored at −20 ◦ C pH of buffers Solubility (␮g/ml)
until analysis. The length of tested segment was measured at the 1.0 1.43 ± 0.03
end of the perfusion. 2.5 1.01 ± 0.02
5.0 1.28 ± 0.04
2.4.2. Perfusion study of AP 5.8 1.36 ± 0.03
6.8 1.56 ± 0.02
The blank perfusion buffer solution was isotonic and composed
7.0 1.63 ± 0.05
of 133 mM NaCl, 4.56 mM KCl, 16 mM NaHCO3 , 1.50 mM NaH2 PO4 7.5 2.16 ± 0.02
and 7.78 mM d-glucose with pH 7.0–7.2 at 37 ◦ C.
Due to the poor solubility of AP in perfusion buffer, Tween 80
was employed to increase the solubility of AP. The perfusate with which was corrected for water flux, was calculated as follows (Li
100 ␮g/ml of AP containing different concentrations of Tween 80 et al., 2011):
(0.5, 1 and 1.5%, w/v) was applied to four intestinal segments to Qout
investigate the effect of Tween 80’s concentration as well as dif- Ccor = Cout ×
Qin
ferent intestinal site on intestinal absorption of AP. To examine
whether intestinal absorption of AP could exhibit concentration- where Cout is the concentration of tested drug in the effluent per-
dependent or concentration-independent characteristics, 10 and fusates, Qin and Qout are the inlet and outlet flow rate (ml/min),
50 ␮g/ml of AP containing 0.5% of Tween 80 were also studied in respectively, which are adjusted for liquid density (ml/min).
all four intestinal segments using the same rat perfusion model. Steady state was reached about 30 min after the beginning of
the perfusion which was confirmed by plotting the ratio of the out-
2.5. Determination of AP by HPLC/UV let to inlet concentrations (corrected for water transport) versus
time. Calculations of permeability across rat intestinal segment
HPLC/UV method was employed for determination of AP in were performed from intestinal perfusate samples collected over
phosphate buffer solution and perfusate on Shimadzu LC-2010AHT 45–120 min (steady state) using the following equation (Li et al.,
HPLC system. AP was separated by an Ultimate C18 column 2011):
(150 mm × 4.6 mm, 5 ␮m) guarded with an Ultimate precolumn −Qin ln (Cout(cor) /Cin )
and detected at 337 nm. Mobile phase consisting of methanol and Peff =
2rl
0.1 mol/l sodium acetate (25/75, v/v) was run at 1.0 ml/min. The
column temperature was set at 30 ◦ C. where Peff is the effective permeability coefficient, Cin is the input
AP was separated well from endogenous materials in perfusate drug concentration, r is the effective lumen radius (cm),
intestinal segments. Within the linear concentration range and l is the length of intestinal segment, 2rl is the area of the
(1.0–120.0 ␮g/ml), good linearity (r2 > 0.9995) was achieved. The mass transfer surface (cm2 ) within the intestinal segment which is
limit of quantification and limit of detection were 10 and 3.3 ng/ml, assumed to be a cylinder area.
respectively. The relative standard deviation (% RSD) of intra-day
and inter-day precision of the assay method for AP was below 5%, 2.7. Statistical analysis
and the accuracy was within the range of 95–105%.
Data were analyzed by two-way ANOVA. Significant difference
2.6. Data analysis between two groups was detected by using LSD test. A probability
level of p < 0.05 was set as the criterion of significance.
2.6.1. Calculation of IDR
To evaluate IDR of AP, the cumulative amount dissolved per sur- 3. Results and discussion
face unit of the compact was plotted against time. The slope of
the linear region (r2 ≥ 0.95) was taken as the intrinsic dissolution 3.1. Solubility and IDR of AP
rate. IDR was calculated by the following equation as described
previously (Zakeri-Milani et al., 2009). The equilibrated solubilities of AP in buffers are presented in
 dw   1  DCs
Table 1. The aqueous solubility of AP over the pH range of 1.0–7.5
IDR = = was quite low with the maximum solubility of 2.16 ␮g/ml observed
dt S h
at pH 7.5. AP is a typical flavonoid with ortho-phenolic dihydroxyl
where IDR is the intrinsic dissolution rate (mg/min/cm2 ); dw is the group at ring A and para-phenolic hydroxyl group at ring B of
change in drug dissolved (mg); dt is the change in time (min); S the flavonoid backbone. The log P value, pKa for first deprotona-
is the surface area of the compact (cm2 ); D is diffusion coefficient tion and water solubility for AP has previously been determined
(cm2 /s); Cs is the solubility (mg/cm3 ) and h is the stagnant layer as 2.87 (Li et al., 1997), 6.6 (Favaro et al., 2007) and 1.35 ␮g/ml
thickness (cm). (Li et al., 1997), respectively, which indicated AP was lipophilic
weak acid and could be ionized at high pH condition. According
2.6.2. Calculation of effective permeability coefficient (Peff ) to Henderson–Hasselbach equation (Wu et al., 2005), AP mainly
Peff was the quantitative estimate of the rate of passage of a presents as hydrophobic molecular form in the tested phosphate
solute across a membrane. It was calculated from the steady-state buffer solutions with pH range of gastrointestinal tract.
concentrations of AP in the perfusate (Cook and Shenoy, 2003). During dissolution test, the concentration of AP was gradually
Because the drug concentrations obtained from the perfusate sam- close to saturate and dissolution rate began to decline at 5 h, which
ples were corrected for changes in the water flux during each time may underestimate the IDR. Therefore, the initial 5 hours’ dissolu-
interval, density corrected gravimetric method was utilized to cal- tion was taken to estimate the IDR (Fig. 2). IDR of AP was calculated
culate the net water flux across the incubated intestinal segment. to be 0.006 mg/min/cm2 . Recently, two studies performed inde-
The density of collected samples was determined by weighing the pendently by Zakeri-Milani et al. (2009) and Yu et al. (2004) on
contents using an electronic weighing balance of a known vol- determining solubility class membership demonstrated good rela-
ume of perfusate. Ccor , the drug concentration of effluent perfusates tionships between IDR and BCS solubility classification. However,
314 J. Zhang et al. / International Journal of Pharmaceutics 436 (2012) 311–317

2.4
The cumulative amount dissolved

2
Y=0.006X+0.453 R =0.996
2.1
per surface(mg/cm )
2

1.8

1.5

1.2

0.9

0.6

0 30 60 90 120 150 180 210 240 270 300 330


T(min)

Fig. 2. Intrinsic dissolution rate (IDR) of AP in 0.05 mol/l phosphate buffer (pH = 6.8).

under the same testing conditions (900 ml of pH 6.8 phosphate


buffer, 100 rpm of rotation speed), almost all of the IDR values Fig. 3. Effects of Tween 80 on the permeability of AP in four intestinal segments.
obtained by Zakeri-Milani et al. were higher than that obtained by
Yu et al. for the same drugs. Such phenomenon could be attributed
separate surfactant concentrations, without the need to conduct
to the different intrinsic dissolution apparatuses and kits used in the
separate free drug concentration determinations. The relationship
two investigations. In case of the stationary disk system employed
between the measured permeability (Papp,uncorr ) in presence of sur-
by Zakeri-Milani et al., a fixed body of solvent was stirred over the
factant, the true permeability (Papp,corr ) in absence of surfactant,
pellet by paddle (USP II method). On the other hand, in case of
micellar association constant (Ka) and the surfactant concentration
the rotation disk system employed by Yu et al., the pellet under-
(Smicellar ) could be described as the following equation:
went a shear-like motion over a planar solvent front, which was
similar to USP Procedure I (Viegas et al., 2001). Different disk sys-
 
1 Ka 1
tems caused different hydrodynamic conditions including fluid = Smicellar × +
Papp,uncorr Papp,corr Papp,corr
velocity and shear rates. In general, USP II generates high fluid
velocity resulting in higher dissolution of tablet compared to USP I A plot of 1/Papp,uncorr versus Smicellar allows estimation of the true
(D’Arcy et al., 2006). Such different stirring situations determined permeability from the Y-intercept (1/Papp,corr ).
the higher IDR values obtained by Zakeri-Milani et al. than that Since Tween 80 showed dose-independent effect on permeabil-
by Yu et al. for the same drugs. Zakeri-Milani et al. and Yu et al. ity of AP in each intestinal segment, the slope of above equation
suggested a borderline value of 1 mg/min/cm2 and 0.1 mg/min/cm2 would be close to zero and the true permeability of AP (Papp,corr )
for determining solubility class membership, respectively. Since we was close to the average Papp,uncorr of AP at three different Tween
followed the experimental procedures of Zakeri-Milani et al. (2009) 80 concentrations.
and the IDR value of AP was much lower than the borderline, AP Historically, surfactants have been utilized to enhance solubility
could be classified as the compound with low solubility. and reduce nonspecific adsorption to the wall of the diffusion
apparatus during the in vitro and in situ permeability estimations
3.2. The effect of concentration of Tween 80 on intestinal of lipophilic compounds (Takahashi et al., 2002; Chen et al., 2010;
permeability of AP Imai et al., 1983; Katneni et al., 2006). However, mechanistic stud-
ies have demonstrated that surfactants may have ability to alter
Since the solubility of AP in buffers (pH 1–7.5) was quite low, membrane integrity (Anderberg and Artursson, 1993), and inhibit
Tween 80 was employed to increase its aqueous solubility. Pre- some transporters (Cornaire et al., 2004; Hugger et al., 2002; Shono
liminary experiments showed no considerable adsorption of the et al., 2004) as well as metabolic enzymes (Christiansen et al.,
compound on the tubing and syringe. The effect of concentration 2011). Although high permeable compounds are easy to across the
of Tween 80 on the intestinal permeability of AP is described in intestinal epithelium, the low solubility of BCS class II compounds
Fig. 3. Increasing concentration of Tween 80 from 0.5 to 1.5% was will limit the concentrations coming into the cells and prevent the
found to result in no significant change in epithelial transport of AP saturation of the efflux transporters, which will affect the rate of
in each intestinal segment (p > 0.05). The dose-independent effect absorption (Wu and Benet, 2005). Comparing with ionic surfac-
of Tween 80 on permeability of each intestinal segment found here tants (e.g. sodium dodecyl sulfate, sodium taurocholate), Tween 80
was consistent to the previous in situ rat perfusion study on ganci- was an ineffective enhancer for some polar drugs (phenol red and
clovir (Li et al., 2011) as well as in vitro Caco-2 transport study on fexofenadine·HCl) due to its ignored local wall damage in intestine
fexofenadine (Lin et al., 2007). The concentration of Tween 80 was (Lin et al., 2007; Katneni et al., 2006; Swenson et al., 1994); while
then set at 0.5% in the perfusate in all other SPIP experiments. comparing to non-ionic surfactant Cremophor-EL, Tween 80 is not
Since the presence of surfactants may either increase or decrease a potent inhibitor of P-gp and incapable of increasing absorptive
the permeability of compound depending on the transporters and transport of digoxin, a typical P-gp substrate (Katneni et al.,
metabolic enzymes involved as well as the integrity of transport 2007). In the way of inhibiting Phase I metabolic enzymes, in vitro
epithelium, it is not possible to estimate the true permeability of study demonstrated that Tween 80 had inhibitory potential on
compound (in absence of surfactants) based upon single concentra- cytochrome P450 (CYP) 3A4-mediated metabolism of testosterone
tion of surfactant used in perfusate. Katneni et al. (2006) developed and the CYP2C9-mediated metabolism of diclofenac with IC50 of
a reciprocal permeability approach for assessment of the true per- 0.40 mM and 0.04 mM, respectively (Christiansen et al., 2011). In
meability by conducting permeability experiments under three addition, Mudra et al. investigated the effects of polyethoxylated
J. Zhang et al. / International Journal of Pharmaceutics 436 (2012) 311–317 315

Table 2
Effective permeabilities of AP in four intestinal segments using single-pass intestinal perfusion.

Perfusate Peff (10−4 cm/s)

Duodenum Jejunum Ileum Colon

AP 10 ␮g/ml 0.645 ± 0.054* 0.377 ± 0.023 0.302 ± 0.042 0.284 ± 0.033


AP 50 ␮g/ml 0.713 ± 0.063* 0.396 ± 0.04N.S,  0.302 ± 0.02N.S,  0.287 ± 0.02N.S, 
AP 100 ␮g/ml 0.502 ± 0.047* 0.198 ± 0.022* ,  0.298 ± 0.02N.S,  0.286 ± 0.03N.S, 

The data were presented as mean ± SD (n = 6 for each group). N.S., not significantly different to the group of 10 ␮g/ml AP in the same intestinal segment.
*
p < 0.05, compared to the other two concentrations of AP in the same intestinal segment.

p < 0.05, compared to the group of duodenum at the same concentrations of AP.

solubilizing agents on drug permeation and metabolism using Fagerholm et al. first investigated the relationship between per-
in situ rat intestinal perfusion model and found Tween 80 imposed meability coefficients in rats and human intestinal, and found that
a great inhibitory effect on the N-demethylation of verapamil the estimated Peff s of passively absorbed compounds in rat jejunum
catalyzed by CYP3A. However, the permeability of parent com- were highly correlated with that in human jejunum (Fagerholm
pound verapamil in presence of Tween 80 was slightly decreased et al., 1996). Later, Salphati et al. (2001) and Zakeri-Milani et al.
compared to the control group (in absence of Tween 80) (Mudra (2007) also confirmed that there was indeed a correlation of effec-
and Borchardt, 2010). For inhibitory of Phase II metabolic enzymes, tive permeability coefficients between rat and human intestinal.
Tween 80 showed mixed-competition type of inhibition on glu- Fagerholm et al. (1996) also predicted the fraction of dose absorbed
curonidation of raloxifene in human liver microsome incubation in human based on the effective permeabilities in human (cal-
(Kim et al., 2009). To the best of our knowledge, in situ and in vivo culated from the permeability determined in rat jejunum), when
evidence of the effects of surfactants on Phase II metabolism was given as a solution or immediate-release dosage form. However, the
still very limited. fraction of dose absorbed in human for AP could not be predicted
from the rat permeability data, since AP has solubility limitation.
3.3. The effect of intestinal site and AP concentration on intestinal In comparison to solubility classification, permeability classi-
permeability fication (especially for the criteria of highly permeability) seems
more difficult, since intestinal permeability is not routinely mea-
Effective permeabilities of AP in four intestinal segments at sured as permeability is based on an absorption measure in humans
three concentrations are presented in Table 2. Intestinal site depen- (Wu and Benet, 2005; Benet et al., 2008). Based on drug metabolism,
dent absorption of AP could be found between duodenum, jejunum Wu and Benet (2005) proposed a new system named “Biopharma-
and ileum/colon at each tested AP concentration. The effective per- ceutics Drug Disposition Classification System (BDDCS)” to provide
meabilities at 10 and 50 ␮g/ml of AP shared the same rank order of a simple surrogate for permeability. Benet et al. (2008) suggested
duodenum > jejunum > ileum ≈ colon, while exhibited another rank that if the extent of drug metabolism (i.e. ≥90%), the drug may be
order of duodenum > ileum ≈ colon > jejunum at 100 ␮g/ml of AP. substituted for 90% absorbed. In other words, if the major route of
The minimum and maximum Peff in rat intestinal segments were drug elimination was metabolism, the drug should be considered
determined to be 0.198 × 10−4 (jejunum) and 0.713 × 10−4 cm/s as high permeable drug; while the drug would be classified as low
(duodenum). The permeability coefficients of AP obtained from permeable drug, when the renal and biliary excretion of unchanged
SPIP study were comparable to the reported values of propranolol drug was the major route of drug elimination. Under BDDCS criteria,
(0.30–0.75 × 10−4 cm/s) (Nagare et al., 2010; Brouwers et al., 2010; the classifications of some drugs changed (i.e. BCS class II talinolol
Varma et al., 2004), which was often used as a reference compound would be BDDCS class IV, since talinolol was poorly metabolized).
of passive absorption via transcellular pathway. In addition, Peff s of Hu et al. (2003) investigated the mechanisms responsible for
AP in the duodenum were significant greater than the other three intestinal disposition of AP in Caco-2 cell culture. During transport
intestinal segments at three AP concentration (p < 0.05), which indi- of AP across the Caco-2 cell monolayer, glucuronidated AP and AP
cated that AP could be well absorbed in the whole intestine with sulfate were biotransformed in the cells, and were subsequently
the main absorption site at duodenum. pumped back to apical side mainly by multidrug resistance-related
Furthermore, there was no statistical difference of Peff s in both proteins (MRPs) and possibly estrone sulfate-sensitive organic
ileum and colon segments at each AP concentration, suggesting the anion transporters (OATs). Such enteric recycling combining with
transport mechanism of AP in such two intestinal segments might enterohepatic recycling led a long-time occurrence of AP in rat
be primarily passive transport. However, significant reductions of blood after oral administration of AP (Gradolatto et al., 2005;
Peff s were observed in the duodenal and jejunum segments (1.4 and Chen et al., 2003). However, similar to many other flavonoids
2.0-fold decrease, respectively), when increasing concentration of as mentioned above, AP was also poor bioavailable due to the
AP from 50 to 100 ␮g/ml. Such concentration-dependent behavior extensive first-pass metabolism related to glucuronidation and
of permeability suggested that AP could be primarily passive trans- sulfation (Chen et al., 2008; Ross and Kasum, 2002). Besides the
port and transcelluar mechanism involving a saturable process, and main metabolic organ liver, intestine may be another potential
potentially via a membrane transport protein in the duodenum and organ responsible for the extensive Phase II metabolism of AP
jejunum (Cook and Shenoy, 2003). (Liu and Hu, 2002; Zhang et al., 2005). Therefore, alternative
In vitro bidirectional transport study of AP on Caco-2 cell mono- route of drug administration would be an effective approach to
layer have been performed by Liu and Hu (2002). Like other enhance the bioavailability of AP by avoiding extensive presystemic
flavonoids (i.e. baicalein (Zhang et al., 2007), wogonin and oroxylin metabolism.
A (Li et al., 2012; Dai et al., 2008)) and isoflavonoids (i.e. genistin
(Liu and Hu, 2002)), the absorptive transport of AP was similar to
its secretory transport with an efflux ratio approaching to 1, indi- 4. Conclusion
cating a passive transport of AP. The determined absorptive Papp
of AP was ∼4.5 × 10−5 cm/s on Caco-2 cell monolayer (Liu and Hu, In the present study, AP was categorized as BCS class II drug due
2002), which suggested an efficient permeability for AP (Volpe et al., to its low solubility and high permeability. The intestinal trans-
2007). port study demonstrated that AP could be absorbed in the whole
316 J. Zhang et al. / International Journal of Pharmaceutics 436 (2012) 311–317

intestine with the main absorption site at duodenum. In addition, Horvathova, K., Novotny, L., Vachalkova, A., 2003. The free radical scavenging activity
the in vivo intestinal absorption mechanism of AP might primar- of four flavonoids determined by the comet assay. Neoplasma 50, 291–295.
Hu, M., Chen, J., Lin, H., 2003. Metabolism of flavonoids via enteric recycling: mech-
ily be passive transport in both ileum and colon segments with anistic studies of disposition of apigenin in the Caco-2 cell culture model. J.
concentration-independent permeability behavior, while might Pharmacol. Exp. Ther. 307, 314–321.
involve both passive and active carrier-mediated transport in both Hugger, E.D., Novak, B.L., Burton, P.S., Audus, K.L., Borchardt, R.T., 2002. A comparison
of commonly used polyethoxylated pharmaceutical excipients on their ability
duodenum and jejunum segments with concentration-dependent to inhibit P-glycoprotein activity in vitro. J. Pharm. Sci. 91, 1991–2002.
permeability behavior. Imai, J., Hayashi, M., Awazu, S., Hanano, M., 1983. Intestinal absorption of dl-
alpha-tocopherol from bile salts and polysorbate 80 micellar solutions in rat.
J. Pharmacobiodyn. 6, 897–902.
Acknowledgements Katneni, K., Charman, S.A., Porter, C.J., 2006. Permeability assessment of poorly
water-soluble compounds under solubilizing conditions: the reciprocal perme-
ability approach. J. Pharm. Sci. 95, 2170–2785.
This research was funded by the Important National Science & Katneni, K., Charman, S.A., Porter, C.J., 2007. Impact of Cremophor-EL and
Technology Specific Projects (No. 2011ZX09201-101-02), Peak of polysorbate-80 on digoxin permeability across rat jejunum: delineation of ther-
six major talents in Jiangsu Province (2010, level A) and Funda- modynamic and transporter related events using the reciprocal permeability
approach. J. Pharm. Sci. 96, 280–293.
mental Research Funds for the Central Universities (Program No. Kim, A.R., Lim, S.J., Lee, B.J., 2009. Metabolic inhibition and kinetics of raloxifene by
JKP2011015). pharmaceutical excipients in human liver microsomes. Int. J. Pharm. 368, 37–44.
Ku, M.S., 2008. Use of the biopharmaceutical classification system in early drug
development. AAPS J. 10, 208–212.
References Li, B., Robinson, D.H., Birt, D.F., 1997. Evaluation of properties of apigenin and [G-3H]
apigenin and analytic method development. J. Pharm. Sci. 86, 721–725.
Amidon, G.L., Lennernäs, H., Shah, V.P., Crison, J.R., 1995. A theoretical basis for a Li, C., Zhang, L., Zhou, L., Wo, S.K., Lin, G., Zuo, Z., 2012. Comparison of intestinal
biopharmaceutic drug classification: the correlation of in vitro drug product absorption and disposition of structurally similar bioactive flavones in Radix
dissolution and in vivo bioavailability. Pharm. Res. 12, 413–420. Scutellariae. AAPS J. 14, 23–34.
Amidon, G.L., Sinko, P.J., Fleisher, D., 1988. Estimating human oral fraction dose Li, M., Si, L., Pan, H., Rabba, A.K., Yan, F., Qiu, J., Li, G., 2011. Excipients enhance intesti-
absorbed – a correlation using rat intestinal-membrane permeability for passive nal absorption of ganciclovir by P-gp inhibition: assessed in vitro by everted gut
and carrier-mediated compounds. Pharm. Res. 5, 651–654. sac and in situ by improved intestinal perfusion. Int. J. Pharm. 403, 37–45.
Anderberg, E.K., Artursson, P., 1993. Epithelial transport of drugs in cell-culture. 8. Lin, H., Gebhardt, M., Bian, S., Kwon, K.A., Shim, C.K., Chung, S.J., Kim, D.D., 2007.
Effects of sodium dodecyl-sulfate on cell-membrane and tight junction perme- Enhancing effect of surfactants on fexofenadine·HCl transport across the human
ability in human intestinal epithelial (Caco-2) cells. J. Pharm. Sci. 82, 392–398. nasal epithelial cell monolayer. Int. J. Pharm. 330, 23–31.
Benet, L.Z., Amidon, G.L., Barends, D.M., Lennernas, H., Polli, J.E., Shah, V.P., Stavchan- Lindenberg, M., Kopp, S., Dressman, J.B., 2004. Classification of orally adminis-
sky, S.A., Yu, L.X., 2008. The use of BDDCS in classifying the permeability of tered drugs on the World Health Organization Model list of Essential Medicines
marketed drugs. Pharm. Res. 25, 483–488. according to the biopharmaceutics classification system. Eur. J. Pharm. Bio-
Brouwers, J., Mols, R., Annaert, P., Augustijns, P., 2010. Validation of a differential pharm. 58, 265–278.
in situ perfusion method with mesenteric blood sampling in rats for intestinal Liu, Y., Hu, M., 2002. Absorption and metabolism of flavonoids in the Caco-2 cell
drug interaction profiling. Biopharm. Drug Dispos. 31, 278–285. culture model and a perfused rat intestinal model. Drug Metab. Dispos. 30,
Chen, J., Lin, H., Hu, M., 2003. Metabolism of flavonoids via enteric recycling: role of 370–377.
intestinal disposition. J. Pharmacol. Exp. Ther. 304, 1228–1235. Logan, C., 2009. Use of animals for the determination of absorption and bioavailabil-
Chen, X., Lu, Y., Du, S., Xu, B., Wang, S., Zhai, Y., Song, X., Li, P., 2010. In situ and in vivo ity. In: Van de Waterbeemd, H., Testa, B. (Eds.), Drug Bioavailability. Wiley-VCH
study of nasal absorption of paeonol in rats. Int. J. Mol. Sci. 11, 4882–4890. Verlag GmbH & Co. KGaA, Germany, pp. 161–184.
Chen, Z.J., Pan, H., Li, L.P., Ting, C., Wang, R.W., Zeng, S., Jiang, H.D., 2008. The oral Mudra, D.R., Borchardt, R.T., 2010. Absorption barriers in the rat intestinal mucosa.
bioavailability of luteolin and apigenin in chrysanthemum morifolium extract 3. Effects of polyethoxylated solubilizing agents on drug permeation and
is much higher than when they are given as pure individual compounds in rats. metabolism. J. Pharm. Sci. 99, 1016–1027.
Drug Metab. Rev. 40, 54–55. Nagare, N., Damre, A., Singh, K.S., Mallurwar, S.R., Iyer, S., Naik, A., Chintamaneni, M.,
Christiansen, A., Backensfeld, T., Denner, K., Weitschies, W., 2011. Effects of non-ionic 2010. Determination of site of absorption of propranolol in rat gut using In situ
surfactants on cytochrome P450-mediated metabolism in vitro. Eur. J. Pharm. single-pass intestinal perfusion. Indian J. Pharm. Sci. 72, 625–629.
Biopharm. 78, 166–172. Papadopoulou, V., Valsami, G., Dokoumetzidis, A., Macheras, P., 2008. Biopharma-
Cook, J., Addicks, W., Wu, Y.H., 2008. Application of the biopharmaceutical classi- ceutical classification systems for new entities (BCS-NMEs) and marketed drugs
fication system in clinical drug development – an industrial view. AAPS J. 10, (BCS-MD): theoretical basis and practical examples. Int. J. Pharm. 361, 70–77.
306–310. Peterson, J., Dwyer, J., 1998. Flavonoids: dietary occurrence and biochemical activity.
Cook, T.J., Shenoy, S.S., 2003. Intestinal permeability of chlorpyrifos using the single- Nutr. Res. 18, 1995–2018.
pass intestinal perfusion method in the rat. Toxicology 184, 125–133. Refsgaard, H.H., Jensen, B.F., Brockhoff, P.B., Padkjaer, S.B., Guldbrandt, M., Chris-
Cornaire, G., Woodley, J., Hermann, P., Cloarec, A., Arellano, C., Houin, G., 2004. Impact tensen, M.S., 2005. In silico prediction of membrane permeability from
of excipients on the absorption of P-glycoprotein substrates in vitro and in vivo. calculated molecular parameters. J. Med. Chem. 48, 805–811.
Int. J. Pharm. 278, 119–131. Rinaki, E., Valsami, G., Macheras, P., 2003. Quantitive biopharmaceutics clas-
Dai, J.Y., Yang, J.L., Li, C., 2008. Transport and metabolism of flavonoids from Chinese sification system: the central role of dose/solubility ratio. Pharm. Res. 20,
herbal remedy Xiaochaihu-tang across human intestinal Caco-2 cell monolayers. 1917–1925.
Acta Pharmacol. Sin. 29, 1086–1093. Ross, J.A., Kasum, C.M., 2002. Dietary flavonoids: bioavailability, metabolic effects,
D’Arcy, D.M., Corrigan, O.I., Healy, A.M., 2006. Evaluation of hydrodynamics in the and safety. Annu. Rev. Nutr. 22, 19–34.
basket dissolution apparatus using computational fluid dynamics – dissolution Salphati, L., Childers, K., Pan, L., Tsutsui, K., Takahashi, L., 2001. Evaluation of a single-
rate implications. Eur. J. Pharm. Sci. 27, 259–267. pass intestinal-perfusion method in rat for the prediction of absorption in man.
Egan, W.J., Lauri, G., 2002. Prediction of intestinal permeability. Adv. Drug Deliv. Rev. J. Pharm. Pharmacol. 53, 1007–1013.
54, 273–289. Shono, Y., Nishihara, H., Matsuda, Y., Furukawa, S., Okada, N., Fujita, T., Yamamoto,
Fagerholm, U., Johansson, M., Lennernas, H., 1996. Comparison between permeabil- A., 2004. Modulation of intestinal P-glycoprotein function by cremophor EL and
ity coefficients in rat and human jejunum. Pharm. Res. 13, 1336–1342. other surfactants by an in vitro diffusion chamber method using the isolated rat
Favaro, G., Clementi, C., Romani, A., Vickackaite, V., 2007. Acidichromism and intestinal membranes. J. Pharm. Sci. 293, 877–885.
ionochromism of luteolin and apigenin, the main components of the naturally Svensson, U.S., Sandstrom, R., Carlborg, O., Lennernas, H., Ashton, M., 1999. High
occurring yellow weld: a spectrophotometric and fluorimetric study. J. Fluoresc. in situ rat intestinal permeability of artemisinin unaffected by multiple dosing
17, 707–714. and with no evidence of P-glycoprotein involvement. Drug Metab. Dispos. 27,
Funakoshi-Tago, M., Nakamura, K., Tago, K., Mashino, T., Kasahara, T., 2011. Anti- 227–232.
inflammatory activity of structurally related flavonoids, apigenin, luteolin and Swenson, E.S., Milisen, W.B., Curatolo, W., 1994. Intestinal permeability enhance-
fisetin. Int. Immunopharmacol. 11, 1150–1159. ment: efficacy, acute local toxicity, and reversibility. Pharm. Res. 11,
Gates, M.A., Tworoger, S.S., Hecht, J.L., De Vivo, I., Rosner, B., Hankinson, S.E., 2007. A 1132–1142.
prospective study of dietary flavonoid intake and incidence of epithelial ovarian Takahashi, T., Kobori, M., Shinmoto, H., Tsushida, T., 1998. Structure-activity
cancer. Int. J. Cancer 121, 2225–2232. relationships of flavonoids and the induction of granulocytic- or monocytic-
Gates, M.A., Vitonis, A.F., Tworoger, S.S., Rosner, B., Titus-Ernstoff, L., Hankinson, S.E., differentiation in HL60 human myeloid leukemia cells. Biosci. Biotechnol.
Cramer, D.W., 2009. Flavonoid intake and ovarian cancer risk in a population- Biochem. 62, 2199–2204.
based case–control study. Int. J. Cancer 124, 1918–1925. Takahashi, Y., Kondo, H., Yasuda, T., Watanabe, T., Kobayashi, S., Yokohama, S., 2002.
Gradolatto, A., Basly, J.P., Berges, R., Teyssier, C., Chagnon, M.C., Siess, M.H., Canivenc- Common solubilizers to estimate the Caco-2 transport of poorly water-soluble
Lavier, M.C., 2005. Pharmacokinetics and metabolism of apigenin in female and drugs. Int. J. Pharm. 246, 85–94.
male rats after a single oral administration. Drug Metab. Dispos. 33, 49–54. Tong, X., Van Dross, R.T., Abu-Yousif, A., Morrison, A.R., Pelling, J.C., 2007. Apigenin
Hillgren, K.M., Kato, A., Borchardt, R.T., 1995. In vitro systems for studying intestinal prevents UVB-induced cyclooxygenase 2 expression: coupled mRNA stabiliza-
drug absorption. Med. Res. Rev. 15, 83–109. tion and translational inhibition. Mol. Cell. Biol. 27, 283–296.
J. Zhang et al. / International Journal of Pharmaceutics 436 (2012) 311–317 317

Ujiki, M.B., Ding, X.Z., Salabat, M.R., Bentrem, D.J., Golkar, L., Milam, B., Talamonti, Wood, J.H., Syarto, J.E., Letterman, H., 1965. Improved holder for intrinsic dissolution
M.S., Bell Jr., R.H., Iwamura, T., Adrian, T.E., 2006. Apigenin inhibits pancreatic rate studies. J. Pharm. Sci. 54, 1068.
cancer cell proliferation through G2/M cell cycle arrest. Mol. Cancer 5, 76–83. Wu, C.-Y., Benet, L.Z., 2005. Predicting drug disposition via application of BCS: trans-
US Food and Drug Administration, 2000. Guidance for Industry, Waiver of In Vivo port/absorption/elimination interplay and development of a biopharmaceutics
Bioavailability and Bioequivalence Studies for Immediate-Release Solid Oral drug disposition classification system. Pharm. Res. 22, 11–23.
Dosage Forms Based on a Biopharmaceutics Classification System. Center for Wu, Z., Razzak, M., Tucker, I.G., Medlicott, N.J., 2005. Physicochemical characteriza-
Drug Evaluation and Research (CDER), Rockville, MD. tion of ricobendazole: I. Solubility, lipophilicity, and ionization characteristics.
Varma, M.V., Panchagnula, R., 2005. Prediction of in vivo intestinal absorption J. Pharm. Sci. 94, 983–993.
enhancement on P-glycoprotein inhibition, from rat in situ permeability. J. Yin, F., Giuliano, A.E., Law, R.E., Van Herle, A.J., 2001. Apigenin inhibits growth and
Pharm. Sci. 94, 1694–1704. induces G2/M arrest by modulating cyclin-CDK regulators and ERK MAP kinase
Varma, M.V.S., Kapoor, N., Sarkar, M., Panchagnula, R., 2004. Simultaneous deter- activation in breast carcinoma cells. Anticancer Res. 21, 413–420.
mination of digoxin and permeability markers in rat in situ intestinal perfusion Yu, L.X., Carlin, A.S., Amidon, G.L., Hussain, A.S., 2004. Feasibility studies of uti-
samples by RP-HPLC. J. Chromatogr. B 813, 347–352. lizing disk intrinsic dissolution rate to classify drugs. Int. J. Pharm. 270,
Viegas, T.X., Curatella, R.U., Winkel Lise, L.V., Brinker, G., 2001. Mesurement of intrin- 221–227.
sic drug dissolution rates using two types of apparatus. Pharm. Technol. 25, Zakeri-Milani, P., Barzegar-Jalali, M., Azimi, M., Valizadeh, H., 2009. Biopharmaceuti-
44–53. cal classification of drugs using intrinsic dissolution rate (IDR) and rat intestinal
Volpe, D.A., Faustino, P.J., Ciavarella, A.B., Asafu-Adjaye, E.B., Ellison, C.D., Yu, L.X., permeability. Eur. J. Pharm. Biopharm. 73, 102–106.
Hussain, A.S., 2007. Classification of drug permeability with a Caco-2 cell mono- Zakeri-Milani, P., Valizadeh, H., Tajerzadeh, H., Azarmi, Y., Islambolchilar, Z., Barze-
layer assay. Clin. Res. Regul. Aff. 24, 39–47. gar, S., Barzegar-Jalali, M., 2007. Predicting human intestinal permeability using
Wang, W., Heideman, L., Chung, C.S., Pelling, J.C., Koehler, K.J., Birt, D.F., 2000. single-pass intestinal perfusion in rat. J. Pharm. Pharm. Sci. 10, 368–379.
Cell-cycle arrest at G2/M and growth inhibition by apigenin in human colon Zhang, L., Lin, G., Chang, Q., Zuo, Z., 2005. Role of intestinal first-pass metabolism of
carcinoma cell lines. Mol. Carcinogen. 28, 102–110. baicalein in its absorption process. Pharm. Res. 22, 1050–1058.
WHO, 2005. Multisource (Generic) Pharmaceutical Products: Guidelines on Zhang, L., Lin, G., KovĂcs, B., Jani, M., Krajcsi, P., Zuo, Z., 2007. Mechanistic study
Registration Requirements to Establish Interchangeability. World Health Orga- on the intestinal absorption and disposition of baicalein. Eur. J. Pharm. Sci. 31,
nization, Geneva. 221–231.

You might also like