Está en la página 1de 10

International Journal of Pharmaceutics 447 (2013) 231240

Contents lists available at SciVerse ScienceDirect

International Journal of Pharmaceutics


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

Enhancement of skin permeation of bufalin by limonene via reservoir type


transdermal patch: Formulation design and biopharmaceutical evaluation
Zhen Yang a,b, , Yang Teng b , Hao Wang b , Huimin Hou b
a
Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, 77030, USA
b
National Pharmaceutical Research Center, Shanghai Institute of Pharmaceutical Industry, Shanghai 201203, China

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

Article history: A reservoir-type transdermal delivery system (TDS) of bufalin was designed and evaluated for various for-
Received 2 December 2012 mulation variables like different penetration enhancers, formulation matrix, rate controlling membranes
Received in revised form 12 February 2013 as well as biopharmaceutical characteristics. Hairless mouse skin was used in permeation experiments
Accepted 22 February 2013
with Franz diffusion cells. In vitro skin permeation study showed that terpenes, especially d-limonene
Available online 1 March 2013
was the most effective enhancer when ethanol and PG were used as the vehicle with a synergistic effect.
Among different rate controlling membranes, ethylene vinyl acetate (EVA) membrane containing 19%
Keywords:
vinyl acetate demonstrated a more suitable release rate for bufalin than the other membranes. In vivo
Transdermal delivery system
Bufalin
pharmacokinetic study of the bufalin patch in rat showed steady-state of bufalin from 3 h to 12 h. In
d-Limonene vivo release rate and cumulative amount analyzed by deconvolution method demonstrated the sus-
Pharmacokinetics tained release of bufalin as long as the patch remained on the animal for at least 12 h. The MRT increased
Deconvolution from 1 h of IV administration to 9 h of transdermal administration. In vitro permeation across mouse
Biopharmaceutics skin was found to have biphasic correlation with plasma AUC in the in vivo pharmacokinetic study.
Current in vitroin vivo correlation (IVIVC) enabled the prediction of pharmacokinetic prole of bufalin
from in vitro permeation results. In conclusion, current reservoir transdermal patch containing 10% d-
limonene as a permeation enhancer, 40% ethanol, 30% PG and 15% carbopolwater gel complex provided
an improved sustained release of bufalin through transdermal administration. The bufalin patch was suc-
cessfully applied to biopharmaceutical study in rats and demonstrated the feasibility of this transdermal
formulation for future development and clinical trials.
2013 Elsevier B.V. All rights reserved.

1. Introduction effects of anti-cancer and pain relief activity, bufalin was approved
as anticancer drugs in China and was used to treat late-stage of
Bufalin is a major component of bufadienolides extracted from liver and lung cancers (Meng et al., 2009; Qin et al., 2008). The
Chinese traditional medicine Chansu (toad venom). It is widely clinical study of bufalin was also conducted in the United States
used as one of the key components in many traditional Chinese for the treatment of pancreatic cancer and showed potential ther-
medicines, i.e., Liu Shen Wan (Hong et al., 1992) and Shexiang apeutic effect (Meng et al., 2009; Wang et al., 2011). Considering
Baoxin Wan (Song et al., 2000). Bufalin exhibits potent anti-tumor low survival rate of liver and lung cancer in late-stage, alternative
activities against liver cancer (Zhang et al., 2012), lung cancer (Zhu medicines obtained from herb materials and animals may provide
et al., 2012), gastric cancer (Li et al., 2009), as well as prostate can- valuable benets.
cer (Yu et al., 2008). Bufalin has been reported to induce apoptosis However, the clinical application of bufalin on cancer treat-
in various cancer cells through both intrinsic and extrinsic path- ment was greatly limited by its unfavorable biopharmaceutical
ways as elaborated in several publications (Hong and Choi, 2012; properties and cardiac toxicity. It has been reported to have poor
Yan et al., 2012; Zhu et al., 2012) and enhance the cytotoxicity of solubility, short half-life and narrow therapeutic window upon
several anticancer drugs (Hashimoto et al., 1997). Because of dual intravenous administration (Yin et al., 2012). The short half-life lim-
its its capability to maintain minimal plasma drug concentration to
achieve therapeutical effect and demands multiple times admin-
istrations. Furthermore, bufalin belongs to toxic steroids, which
produce digoxin-like cardiac toxicity (Wang et al., 1997). Bufalin
Corresponding author at: Department of Pharmacological and Pharmaceutical
has side effect of cardiac arrhythmia, breathlessness, convulsion
Science, College of Pharmacy, University of Houston, 77030, USA.
and coma at high dosage, which may cause safety issues (Panesar,
Tel.: +1 832 403 6299.
E-mail address: zyang9@uh.edu (Z. Yang).
1992).

0378-5173/$ see front matter 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.ijpharm.2013.02.048
232 Z. Yang et al. / International Journal of Pharmaceutics 447 (2013) 231240

Fig. 1. The chemical structure of bufalin.

Currently developed formulation of bufalin include wheat


germ agglutinin-grafted lipid nanoparticles (Liu et al., 2011), self-
microemulsing lipid-based formulation (Liu et al., 2010) and
Fig. 2. The schematic preparation of reservoir-type bufalin patch.
PEGPLGA loaded nanoparticles (Yin et al., 2012). Although they
have achieved some successes on improving solubility and overall
bioavailability compared to traditional tablet and injections (Yang 3MTM CoTranTM 9728, 3MTM CoTranTM 9705, 3MTM CoTranTM 9715
et al., 2008), the major issues including high dose derived car- and other types of ethylene vinyl acetate (EVA) membrane with dif-
diotoxicity and rapid decline of exposure still exist. Transdermal ferent vinyl acetate content 2%, 9%, 19%, 28% were obtained from
formulation was considered as an enabling strategy for this sce- 3M Pharmaceuticals (St. Paul, MN, USA). Diphenhydramine, oleic
nario by offering both controlled and sustained release patterns. A acid, lauric acid, sodium lauryl sulphate (SLS), azone, ethyl oleate,
number of FDA approved transdermal products, like Transderm- ethyl lauric acid, isopropyl myristate (IPM), Tween 80, N-methyl-
NitroTM , EstradermTM , DuragesicTM , D-TRANSTM , AndrodermTM 2-pyrrolidone (NMP), ethanol, propylene glycol (PG), 1,8-cineole,
and Catapres-TTSTM had demonstrated the successful application d-limonene and l-menthol were purchased from Sigma (St. Louis,
of transdermal administration. MO, USA). Carbopol 934 P (pharmaceutical grade) was purchased
To the best of our knowledge, there is no transdermal formu- from Goodrich Co. Ltd., (Cleveland, OH, USA). All other chemicals
lation of bufalin that has been developed and evaluated. Bufalin were obtained from Shanghai Chemical Reagents Co. (Shanghai,
is a small molecular compound (molecular weight = 386.5) with a China). Aluminum foilpolyethylene backing layer was purchased
suitable lipophilicity (log P = 2.78). However, its poor aqueous sol- from Shanghai Package Material Company (Shanghai, China). Heat-
ubility (33 g/ml) may limit permeation force through dermis (Liu sealing machine was purchased from Yiheng Inc. (Shanghai, China).
and Feng, 2008). The chemical structure of bufalin is shown in Fig. 1. Transdermal permeation diffusion instrument was purchased from
For drugs with a short biological half-life and/or a narrow thera- Jiekai Inc. (Shanghai, China).
peutical range, transdermal administration is one of the attracting
alternative approaches to decrease toxicity and prolong therapeu- 2.2. Preparation of transdermal delivery system (TDS)
tical action. Therefore, the skin has become an important route for
drug delivery for topical, regional or systemic effects for the last Reservoir-type TDS was prepared with an active membrane
two decade (Paudel et al., 2010). The major challenges of develop- release area of 15 cm2 . The schematic of TDS preparation in the
ing transdermal drug delivery lie on the poor permeation of API and present study is shown in Fig. 2.
its potential skin irritation. Using chemical enhancers is the most
common strategy to improve the permeation of drugs through the 2.2.1. Preparation of transdermal gel of bufalin
skin barrier. 40 mg bufalin was rstly fully dissolved in 4 ml ethanol, and then
Therefore, the objective of this study can be divided into 3 ml PG added. 1 ml d-limonene (10%, w/v of mixture) was added
three aims, (1) develop a transdermal delivery system for bufalin as a penetration enhancer to drug solutions, and then 0.1 ml Tween
administration, (2) evaluate its biopharmaceutical characteristics 80 added as a surfactant to form a uniform phase. 1 g carbopol used
to ensure sustained release of bufalin in vivo, (3) perform in vitroin as gelling agent was swelled in 50 ml double distilled water (form
vivo correlation and use in vitro data to predict and guide pharma- 2% carbopol gel) overnight while pH value was adjusted to 7.4. 1.5 g
cokinetics in future. carbopolwater gel (2%) was added to drug solutions (15%, w/v) and
stirred well to form a uniform gel. The uniformity of drug content
2. Materials and methods (4 mg/ml) was tested for all batches of gels by HPLC.

2.1. Materials 2.2.2. Preparation of reservoir transdermal patch


A procedure for drug-loaded-membrane laminate was divided
Bufalin was extracted and puried in our lab from Traditional into three main phases. Phase 1 involved lling of drug reservoir.
Chinese Medicine, Chansu. The puried bufalin powder had been 1 ml bufalin gel was squeezed to the EVA membrane from peri-
identied by NMR and Mass Spectrometry and quantied by HPLC staltic pump and then the backing layer was used to cover the
with a purity >98% in our previous study (Yang et al., 2008). rate-controlling membrane on the top of the druggel complex. The
Z. Yang et al. / International Journal of Pharmaceutics 447 (2013) 231240 233

drug content uniformity was ensured to be within 5% of total dose 2.5. In vivo pharmacokinetic study in rats
(4 mg/ml). Phase 2 involved heat-sealing of the backing layer using
the heating-sealing machine. The backing layer was placed on the The animal protocols used in this study were approved by Insti-
top of the reservoir, and the heater which was preheated at 90 C tutional Animal Care and Uses Committee from Shanghai Institute
was forced down pneumatically for 10 s to form a reservoir patch of Pharmaceutical Industry. Male Sprague-Dawley rats weighing
with 15 cm2 (3 cm 5 cm). Phase 3 involved completing patch with 250 20 g were used for pharmacokinetic studies. Each pharma-
the adhesive layer. The reservoir was placed on the adhesive layer cokinetic study was performed on three rats and blood samples
with EVA membrane upside and covered with anti-stick liner. An (200250 l) were collected via retro-orbital plexus using a ster-
entire bufalin patch is shown at the end of Fig. 2. ilized glass capillary tube. For i.v. bolus pharmacokinetic study,
a dose of 100 g/ml bufalin was prepared in 10% alcohol saline
2.3. In vitro skin permeation studies solution. The injection solution was ltered through the 0.45 m
membrane and sterilized at 115 C for 30 min in a sealed ampoule.
The dorsal skins of hairless mice (2025 g) were excised after Blood samples were drawn at 0.083, 0.167, 0.333, 0.667, 1, 2, 4, 6
sacrice by cervical dislocation. Fresh prepared skins were stored in and 8 h following the intravenous administration. For transdermal
a refrigerator at 20 C without repeatable freeze and thaw cycles. administration, bufalin patches (3 cm 5 cm, 4 mg) were applied to
The storage of skin was limited within 1 month otherwise new the shaved abdomen of rats. Rats were carefully shaved by electric
batch of skin would be prepared. Prior to permeation experiments, shaver one day before transdermal administration. The blood sam-
skin was thawed and subcutaneous fat, tissue and capillaries of ples were collected at 0.5, 1, 2, 4, 6, 8, 10, 12, 13, 14 and 24 h while
skin were carefully removed. After cutting into pieces, skin was the patches remained on the animal up to 12 h. The blood samples
mounted between the donor and receptor compartments of the were immediately centrifuged at 10,000 rpm for 10 min, and the
Franz diffusion cells with the stratum corneum facing the donor plasma was separated and stored at 20 C until analysis.
compartment.
The permeation area of Franz diffusion cells was 2.84 cm2 and 2.6. Pharmacokinetic and deconvolution analysis
a receiver volume was 7.0 ml. 40% PEG and 30% PG aqueous solu-
tion were used as the receiver medium and 50% PG solution was WinNonlin 5.3 (Pharsight Corporation, Mountain View, CA)
used as donor solution with 4 mg/ml bufalin. Assembled diffusion was used for bufalin pharmacokinetic analysis. Both compartment
cells in triplicate were placed in a transdermal permeation diffu- and noncompartmental model were applied for the pharmacoki-
sion instrument and maintained isothermally at 37 C. The receptor netic analysis of bufalin proles. IV pharmacokinetic analysis was
compartment was stirred with a magnetic stirrer at 250 rpm. Care performed on compartmental model and transdermal administra-
was taken to ensure that no air bubbles remained in the receiver tion was conducted on noncompartmental model. Pharmacokinetic
side. Samples (0.1 ml) were withdrawn at 0.5, 1, 2, 4, 6, 8 and 10 h parameters, including Cmax , Tmax , half-life, mean residence time
for HPLC analysis and were replaced with an equivalent volume. To (MRT), CL, Vss , AUC and AUMC were derived directly from WinNon-
minimize evaporation, the donor compartment was covered with lin. Deconvolution analysis was also performed to estimate in vivo
paralm and aluminum foil. bufalin release from transdermal batch (Sun et al., 2012). IV does
In vitro permeation prole of transdermal patch was performed pharmacokinetics (100 g) was used as a reference formulation
to correlate with in vivo pharmacokinetic behavior. Intact bufalin for an impulse-response function. Pharmacokinetics of transder-
patches were mounted on Franz diffusion cells across hairless mal patch was analyzed by deconvolution methodology to obtain
mouse skin and the experiment procedure is similar to in vitro skin input-response function, i.e., in vivo release rate and cumulative
permeation study. release amount. The number of predicted values was 200 and iter-
ation number was 100. Maximum number of UIR (Unit Impulse
2.4. Data analysis Response) exponentials was set at 3 and model selection was based
on Akaike Information Criteria (AIC) (Bozdogan, 2000).
The cumulative amount (Q, g/cm2 ) of bufalin permeated
through skin was calculated by the following equation. 2.7. In vitroin vivo correlation

n1 The cumulative amount of bufalin permeated across mouse skin


Cn V + 0.1 i=1
Ci
Q = (1) in vitro from bufalin batch was compared against the extent of
S
absorption and cumulative AUC values to establish in vitroin vivo
where S is the effective area, V is the volume of receptor cell, Cn correlation (IVIVC). This correlation represents a point-to-point
is the bufalin concentration at time point n, and Ci is the bufalin relationship with the same time point (Type A).
concentration at time point i.
The cumulative amount (g/cm2 ) of bufalin permeated through 2.8. Sample preparation and quantitative determination of
skin was plotted versus time (h). Each data is the mean SD of three bufalin
determinations. JSS represents the best-t slopes of the apparent
linear portions, usually several data points in the beginning were Analysis of bufalin in rat plasma was performed by API 3000
removed during tting. KP is the apparent permeability coefcient triple-quadrupole mass spectrometer coupled with Shimazdu LC-
and is calculated according to Ficks rst law of diffusion, 20 AD HPLC system. The quantication was performed in a
JSS positive ion mode using multiple reaction monitoring (MRM) with
KP = (2) 387.4/351.2 (m/z) for bufalin and 256.2/167.0 (m/z) for diphen-
Co
hydramine (Internal standard). The other instrument dependent
where JSS is steady state ux and Co is the initial drug concentration parameters were as following: ionization gas, 11 L/min; curtain
in donor side. Tlag was determined by extrapolating the straight- gas, 10 L/min; collision gas, 7 L/min; ion voltage, 4500 V; ion tem-
line portion of each steady-state permeation curve to the time axis. perature, 400 C. The HPLC conditions for analyzing bufalin in rats
The ux data were subjected to student T-test and one-way analysis plasma: analytical column, C18 Diamonsil (50 mm 2.1 mm I.D.
of variance (ANOVA) to determine the level of signicance. The data 5 m), and the mobile phase was isocratic using methanolwater
was considered to be signicant at P < 0.05. (70:30, 5 mM ammonium acetate) with a ow rate of 0.2 ml/min.
234 Z. Yang et al. / International Journal of Pharmaceutics 447 (2013) 231240
Cumulative amounts of permeated bufalin

Cumulative amounts of permeated bufalin


4.0 oleic acid D-li monene no enhancer 40
PG/water (50/50)
lauric acid 1,8-cineole
3.5 PG/PEG400/water (10/40/50)
IPM/NMP L-menthol
3.0 azone ethyl lauric acid 30 ethanol/PG/water (40/30/30)

2.5 SLS ethyl oleate


(g/cm2)

(g/cm 2)
2.0 20
1.5
1.0 10
0.5
0.0
0 2 4 6 8 10 12 0
0 2 4 6 8 10 12
Time (h) Time (h)
Fig. 3. Effect of skin permeation enhancers (5%, w/v) on the permeation of bufalin
Fig. 4. Permeation proles of bufalin in different vehicle solutions. Data were pre-
in PG/water cosolvent system (n = 3).
sented as the mean SD (n = 3).

The chromatography was performed at 30 C and the injection


signicantly increased the permeation rate of bufalin from 0.021
volume was 5 l. Analysis of bufalin in permeation samples was
to 0.437 g/cm2 /h (22-fold). Permeation parameters of bufalin
performed by reversed phase HPLC using a Shimazdu LC-10ADvp
with 5% (w/v) of various enhancers across hairless mouse skin at
HPLC system, which was validated in our previous publication
37 C are shown in Table 1.
(Yang et al., 2008). LCMS/MS method validation including intra-
and inter-day variation, stability, linearity and matrix effect were
performed with the same methodology shown before (Yang et al., 3.2. Effect of vehicle selection on the skin permeation of bufalin
2010).
A volume of 25 l of methanol (containing 200 ng/ml diphenhy- In order to investigate the synergistic effect of different
dramine) was added to 100 l aliquot of rat plasma samples. 1 ml vehicles on the bufalin permeation with d-limonene, the accu-
ether was added to extract and vertex for 5 min. After centrifuge at mulated amount of bufalin permeated across hairless mouse skin
3000 rpm for 10 min, the supernatant was evaporated to dryness at from saturated solutions with PG/PEG 400/water (10/40/50, v/v),
40 C under nitrogen and reconstituted in 100 l of mobile phase. A ethanol/PG/water (40/30/30, v/v) and PG/water (50/50, v/v) is
5 l portion of each sample was injected into the LCMS/MS system shown in Fig. 4. Ethanol/PG/water (40/30/30, v/v) showed 9 times
for analysis. higher accumulative permeated amount of bufalin compared to
PG/water (50/50, v/v) (P < 0.05). Permeation parameters of bufalin
2.9. Skin irritation across hairless mouse skin with different vehicle solutions at 37 C
are shown in Table 1.
Evaluation of skin irritation was performed by visual obser-
vation after pharmacokinetic and pharmacodynamic studies in 3.3. Effect of enhancer concentration on the skin permeation of
Sprague-Dawley rats. Histological evaluations will be performed bufalin
if obvious redness and swelling are observed.
To estimate the enhancement of the different concentrations
3. Results of d-limonene, various concentrations (5%, 7%, 10%, 15%) were
added to the ethanol/PG/water (40/30/30) cosolvent system. Per-
3.1. Effect of permeation enhancers on the skin permeation of meation of bufalin increased proportionally as the concentration
bufalin of d-limonene increased from 5% up to 10% (Fig. 5). The results

The control study using nicotine as a model compound was


Cumulative amounts of permeated bufalin

75 5%D-limonene
performed and ux results were similar to those published in liter-
ature (Zorin et al., 1999). Except for a short lag time in the beginning 7%D-limonene
of permeation study, the cumulative permeation amount of bufalin 10%D-limonene
proportionally increased with time up to 10 h, indicating a good
50 15%D-limonene
integrity of skin during permeation study. Bufalin showed an
(g/cm2)

intrinsic ux of 21 ng/cm2 /h across hairless mouse skin when 50%


PG was used in donor side. The low permeation rate indicated
that chemical enhancers were needed for transdermal admin-
25
istration. The feasibility of using oleic acid, lauric acid, sodium
lauryl sulphate, ethyl oleate, ethyl lauric acid, isopropyl myristate,
N-methyl-2-pyrrolidone, azone, 1,8-cineole, d-limonene and
l-menthol to reduce permeation barrier was investigated. The
0
enhancement on the skin permeation rate of bufalin by adding 5%
0 2 4 6 8 10 12
(w/v) of various enhancers to the cosolvent system is shown in
Fig. 3. Compared to terpenes including d-limonene, l-menthol and Time (h)
1,8-cineole, other types of chemical enhancers increased the ux
Fig. 5. Effect of d-limonene concentration (%, w/v) on the permeation of bufalin
of bufalin within 5-fold. Terpenes, especially d-limonene, were saturated in ethanol/PG/water (40/30/30) cosolvent system. Data were presented
the most effective among ten different chemical enhancers, which as the mean SD (n = 3).
Z. Yang et al. / International Journal of Pharmaceutics 447 (2013) 231240 235

Table 1
Permeation parameters of bufalin through excised hairless mouse skin from 4 mg/ml bufalin solution with different formulation variables at 37 C (n = 3).

Formulation # Membrane Carbopol Matrix Enhancer JSS (g/cm2 /h) KP (103 , cm/h) Tlag (h) ER
gel

1 PG/water (50/50) Control 0.02 0.01 0.01 0 1.92 0.42


2 PG/water (50/50) 5% oleic acid 0.10 0.01* 0.02 0.0* 2.32 0.35 5.1
3 PG/water (50/50) 5% lauric acid 0.11 0.04* 0.04 0.01* 2.89 0.07 5.2
4 PG/water (50/50) 5% sodium lauryl sulfate 0.05 0.01 0.01 0.0 3.97 0.21* 2.5
5 PG/water (50/50) 5% azone 0.05 0.01 0.02 0.0 2.76 0.19 2.4
6 PG/water (50/50) 5% ethyl oleate 0.03 0.0 0.01 0 2.27 0.23 1.4
7 PG/water (50/50) 5% ethyl lauric acid 0.11 0.04* 0.03 0.01* 2.25 0.03 5.3
8 PG/water (50/50) 5% IPM/NMP 0.04 0.01 0.01 0.0 2.63 0.26 2.0
9 PG/water (50/50) 5% 1,8-cineole 0.35 0.04* 0.08 0.01* 4.08 0.25* 17.1
10 Ethanol/PG/water (40/30/30) 5% d-limonene 3.86 0.53* 0.99 0.06* 2.86 0.21 183.5
11 PG/PEG400/water (10/40/50) 5% d-limonene 0.59 0.12* 0.15 0.01 1.94 0.32 28.1
12 PG/water (50/50) 5% d-limonene 0.44 0.09** 0.11 0.01* 4.11 0.46* 22.2
13 Ethanol/PG/water (40/30/30) 7% d-limonene 5.82 0.54** 1.32 0.09* 2.95 0.32 277.6
14 Ethanol/PG/water (40/30/30) 10% d-limonene 8.73 1.35** 1.57 0.17* 2.04 0.43 415.7
15 Ethanol/PG/water (40/30/30) 15% d-limonene 8.92 1.54** 1.64 0.21* 2.32 0.37 424.7
16 PG/water (50/50) 5% l-menthol 0.32 0.06* 0.08 0.0* 4.16 0.38* 15.3
17 15% Ethanol/PG/water (40/30/30) 10% d-limonene 6.84 1.14** 1.44 0.25* 4.02 0.27* 325.7
18 25% Ethanol/PG/water (40/30/30) 10% d-limonene 3.62 0.56** 0.99 0.16* 4.16 0.31* 172.3
19 40% Ethanol/PG/water (40/30/30) 10% d-limonene 0.48 0.14* 0.12 0.03* 4.24 0.32* 24.0
20 19% EVA 15% Ethanol/PG/water (40/30/30) 10% d-limonene 5.66 1.01** 1.33 0.20* 4.80 0.42* 269.3

Note: Donor side concentration of bufalin was assumed as 4 mg/ml in various donor solutions to facilitate calculation and they could be supersaturated solutions.
Data are the means SD of three independent experiments.
ER is the enhancement ratio calculated by JSS of treatment versus control.
*
P < 0.05 compared to control group.
**
P < 0.01 compared to control group.

showed the permeation enhancement is concentration-dependent, 3.5. Effect of gelling agent


and achieved the highest permeation effect at 10%. As no signicant
difference on the effects of permeation enhancement between 10% In order to increase the viscosity of the reservoir solution to facil-
and 15%, 10% d-limonene was chosen as the appropriate concentra- itate manufacture, Carbopol 934 P was added in the solution since
tion of enhancer. Permeation parameters of bufalin across excised it is known as a thickening agent or viscosity modier, which has
hairless mouse skin with various concentration of d-limonene at been mainly used in gels, suspensions and emulsions. After dissolv-
37 C are shown in Table 1. ing bufalin in ethanol/PG (40/30, v/v) solution with 10% d-limonene
and 1% Tween 80, the effect of different volumes of carbopol gel on
3.4. Effect of membrane on the controlled release of bufalin the permeation proles across the EVA membrane/skin compos-
ite was studied. As shown in Fig. 7, the permeation rate of bufalin
To control the release of bufalin from the reservoir com- decreased from 6.84 to 0.48 g/cm2 /h when the concentration of
partment, EVA copolymer membrane was selected as the Carbopol 934 P increased from 15% to 40% (w/v). When the content
rate-controlling membrane. EVA copolymer membranes used in of gelling polymer is less than 15%, the viscosity of bufalin solution
current study contained various content of vinyl acetate [2%, 9%, is not sufcient to remain in the center of backing layer during heat
19%, 28% (w/w)] with the uniform thickness of 76.20 m. When sealing. The results showed the addition of 15% (v/v) of carbopol
bufalin was dissolved in ethanol/PG/water (40/30/30) containing gel provided desirable rheological properties with the insignicant
10% (w/v) d-limonene, the permeation proles of bufalin across reduction in bufalin permeation rate.
the EVA copolymer membrane are shown in Fig. 6. The membrane
permeation rate of bufalin increased as the content of vinyl acetate
increased from 2% up to 28%.
Cumulative amounts of permeated bufalin

200
Cumulative amounts of permeated bufalin

2%vinyl acetate 19%vinyl acetate


45 15% carbopo l gel
40 25% carbopo l gel
9%vinyl acetate 28%vinyl acetate
150 35
40% carbopo l gel
30
(g/cm2)
(g/cm2)

25
100
20
15
50 10
5
0 0
0 2 4 6 8 10 12 0 2 4 6 8 10 12
Time (h) Time (h)

Fig. 6. Permeation proles of bufalin across the EVA membrane with various content Fig. 7. Permeation proles of bufalin through membrane/skin composite with var-
of vinyl acetate. Data were expressed as the mean SD (n = 3). ious volume of carbopol gel. Data were expressed as the mean SD (n = 3).
236 Z. Yang et al. / International Journal of Pharmaceutics 447 (2013) 231240

Table 3
Cumulative amounts of permeated bufalin
45 Pharmacokinetic parameters of bufalin after IV and transdermal administration in
no adhesive layer rats (n = 3).
40
with adhesive layer Parameters Transdermal IV
35
30 Cmax (ng/ml) 174.77 45.24 657.81 125.43
Tmax (h) 8.0 2.0
(g/cm2)

25 MRT (h) 9.17 1.05 1.09 0.09


T1/2 (h) 3.15 0.63 0.76 0.06
20 Cl (ml/h) 143.63 35.78
15 Vss (ml) 156.02 31.62
AUC0 (h ng/ml) 2093.66 566.46 722.38 157.41
10 AUMC0 (h h ng/ml) 19,190.66 5848.01 796.28 210.79
5 Data are the means SD of three rats.

0
0 2 4 6 8 10 12
Time (h) aqueous samples. There was no interference in rat plasma regarding
to determination of bufalin, and the chromatograms are shown in
Fig. 8. Permeation proles of bufalin through membrane/skin composite with poly- Supplemental Fig. 1.
acrylic acid pressure sensitive adhesive. Data were presented as the mean SD
(n = 3).

3.8. Pharmacokinetic and deconvolution analysis of bufalin patch


3.6. Effect of pressure sensitive adhesive on the permeation of in rats
bufalin
The pharmacokinetic studies were conducted in rats for intra-
The EVA membrane containing 19% vinyl acetate was chosen as venous (100 g/ml) and transdermal routes (4 mg/ml). The mean
controlled release membrane, and pressure sensitive adhesive con- plasma concentrationtime of bufalin after IV and transdermal
taining polyacrylic acid (total contents = 55%) was coated with the administration is presented in Fig. 9. A summary of the pharmacoki-
thickness of 5.0 m. Permeation proles of bufalin through the EVA netic parameters is shown in Table 3. Pharmacokinetic parameters
membrane coated with and without pressure sensitive adhesive are for IV administration were tted by both one and two compartment
shown in Fig. 8. Pressure sensitive adhesive with polyacrylic acid models. As seen in Fig. 9A, bufalin did not differentiate distribution
was usually used as the adhesive agent to keep the close contact phase and elimination phase obviously after IV administration and
between the patch and skin. The permeation rate of bufalin through the concentration at 8 h was below detection limit. Based on AIC
the adhesive layer was much lower than that without the adhesive value, one compartment model showed smaller AIC values (60)
layer (only 15% of the original rate). This may be due to the interac- than two compartment models (90), therefore one compartment
tion between the carbopol gel and polyacrylic acid, which resulted model was used to estimate parameters for IV administration.
in the barrier between the adhesive layer and skin. Therefore, pres- IV dose of bufalin showed that bufalin has a very short half life
sure sensitive adhesive (polyacrylic acid type) was coated on the (0.8 h) and MRT (1 h). The clearance was estimated at 143.63 ml/h
peripheral region of the patch to avoid decreasing the skin per- and steady-state volume distribution was 156 ml. For transder-
meation of bufalin. The nal formulation composition is shown in mal administration, non-compartment model was used. In vivo
Table 2. pharmacokinetic study showed steady state around 3 h. MRT of
transdermal administration (9.2 h) is much longer than IV admin-
istration (1.1 h). The plasma concentrations of bufalin remained
3.7. Analytical method validation
above 100 ng/ml from 3 h until patch was removed at 12 h.
Deconvolution analysis showed that patch was able to provide
The method validation was conducted in blank rat plasma. The
sustained release of bufalin from 2 h up to 12 h. The estimated
standard curves of bufalin were linear in the concentration range
release rate achieved peak at 4 h and it was consistent with the
from 2.5 to 1000 ng/ml, and the lower limit of quantitation was
lag time shown at in vitro permeation study (lag time = 4 h). After
2.5 ng/ml in rat plasma. 1/x2 was used a weighting factor to quantify
8 h, the estimated release rate gradually declined but still main-
bufalin plasma concentration. Linear regression showed R2 value
tained at a relatively stable level (Fig. 9B). The predicted in vivo
equals to 0.999. The intraday and interday precisions of QC samples
release amounts showed almost linear increase after 2 h transder-
at three concentration levels (20, 200, and 1000 ng/ml) are shown
mal administration (Fig. 9C), which was also consistent with both
in Supplemental Table 1. Rat plasma samples containing bufalin
in vitro and in vivo behaviors of the bufalin batch. Both pharmacoki-
showed within 15% CV in short-term (8 h at 20 C), long-term (4
netic and deconvolution results demonstrated that reservoir type
weeks at 20 C) and three freezethaw cycle stability study. No
transdermal patch of bufalin signicantly improve the sustained
obvious matrix effect was detected in plasma samples compared to
release of bufalin in vivo and the decreased dose frequency.

Table 2
Formulation composition of bufalin transdermal patch (%, w/v). 3.9. In vitroin vivo correlation
Components Contents
IVIVC between the cumulative % of bufalin permeated across
Bufalin 4 mg
mouse dorsal skin and plasma AUC at the same time point showed
Carbopol gel 15%
Ethanol 40%
a biphasic curve pattern as shown in Fig. 10, which can be clearly
PG 30% distinguished into two stages. Good linear correlation was observed
d-Limonene 10% with correlation coefcients, R2 = 0.999 during the initial phase
Tween-80 1% (Fig. 10B) and R2 = 0.972 during steady-state (Fig. 10C). The mean
Water 4%
AUC of bufalin in vivo was in agreement with the observed perme-
The total volume of bufalin transdermal patch is 1 ml. ation prole in vitro.
Z. Yang et al. / International Journal of Pharmaceutics 447 (2013) 231240 237

B
40
35

In vivo release rate (g/h)


30
A I.V. 25
2000 Patch 20
Plasma Concentraons (ng/ml) 15
Remove the
patch at 12 h 10
200 5
0
0 2 4 6 8 10 12
20 Time (h)

C 250

In vivo cumulave release


2 200
0 4 8 12 16 20 24

amounts (g)
Time (h)
150

100

50

0
0 2 4 6 8 10 12
Time (h)

Fig. 9. Plasma concentrationtime proles of bufalin after IV and transdermal administration. Data are presented as mean SD. The doses for intravenous and transdermal
routes were 100 g and 4 mg, respectively.

3.10. Skin irritation Sprague-Dawley rats. No obvious redness and swelling were found
on skin after 12 h patch administration. Bufalin was not thought
Evaluation of skin irritation was performed by visual obser- to be a skin irritant based on present transdermal patch study in
vation after pharmacokinetic and pharmacodynamic studies in animal model.

A 1600

1200
AUC (ng/ml*hr)

800

400

0
0 200 400 600 800

Amount permeated (g) in vitro

B 120 C 2000

100 y = 2.481x + 1.200 y = 2.030x + 307.6


1600
R = 0.998 R = 0.972
AUC (ng/ml*hr)

AUC (ng/ml*hr)

80
1200
60
800
40

20 400

0 0
0 10 20 30 40 0 200 400 600 800
Amount permeated (ug) in vitro Amount permeated (ug) in vitro

Fig. 10. In vitroin vivo correlation of cumulative amount permeated in vitro versus plasma AUC in Sprague-Dawley rats (n = 3). It is a type A IVIVC (point to point).
238 Z. Yang et al. / International Journal of Pharmaceutics 447 (2013) 231240

4. Discussion PG, thus increasing concentration gradient of permeation between


donor compartment and receiver compartment. Secondly, per-
This is the rst systematic study of development and biophar- meation of ethanol into the stratum corneum can alter the
maceutical evaluation of bufalin via transdermal administration. solubility properties of the tissue with a consequent improve-
Transdermal formulation as an alternative delivery route for ment for drug partitioning into the membrane. Additionally,
bufalin is proposed based on the following perspectives. Firstly, ethanol may extract some of the lipid fraction within stratum
bufalin is well known for its cardiotoxicity, and current formula- corneum when used at high concentration for prolonged times,
tions, i.e., tablet and injection, usually cause serious side effect. and thus improve drug ux through skin (Krishnaiah et al.,
Transdermal formulation provides constant and controlled release 2002).
of drug which may solve this issue and keep plasma concentration When the analyte is lipophilic which has low solubility in the
in the desired therapeutic window. Secondly, physicochemical receiver side, modifying of the receptor uid is recommended in
properties of bufalin indicate that it might be suitable for transder- order to achieve accurate determination of permeated amounts.
mal administration when its molecular weight is less than 500 and In our current study, PEG/PG/water (40/30/30, w/v) was used as
log P value is around 3. Thirdly, bufalin is a potent therapeutical receiver medium and permeated bufalin was completely dissolved
agent which makes transdermal formulation viable due to low in the receiver side for up to 10 h. In order to keep sink conditions
dose requirement. It showed strong induction effect on differenti- to maintain a favorable driving force for permeation, 4 mg/ml was
ation of human leukemia at as low as 10 nM (Kawazoe et al., 1999; chosen as the donor side concentration, the same dose as in vivo
Zhang et al., 1992). Other reports showed that bufalin effectively pharmacokinetic study. The permeated bufalin was less than 20%
inhibits proliferation of gastric, prostate cancer cells and other of total amount in the donor side after 12 h permeation study and
cancer cells within the range of several hundred nanomolar (Dong the concentration of bufalin in the receptor uid was always below
et al., 2011; Li et al., 2009; Yeh et al., 2003). Our pharmacodynamic 5% of its concentration in the donor medium.
study also showed no signicant cardiotoxicity of bufalin up to The permeation rate of bufalin across 28% vinyl acetate EVA
1500 nM of plasma concentration in rodents (data not shown). membrane was higher than its permeation rate across skin
Therefore, the current bufalin plasma concentration is perfectly (Figs. 5 and 6). In order to design a device controlled transdermal
tted in the therapeutic window. patch formulation, EVA membrane with 19% of vinyl acetate was
Although other chemical enhancers also increased permeation chosen as the controlled release membrane.
of bufalin, the chemical enhancer screening result implied that d- In the present study, hairless mouse skin was used as in vitro
limonene is the most potent enhancer by increasing 22 times higher permeation model and rats were used at in vivo pharmacokinetic
permeation rate of bufalin than control. As a result of the synergy study. These models were chosen because other cardiotoxicity and
between d-limonene and ethanol, the enhancement was greater anti-cancer activity studies of bufalin were performed on rodents,
than 400 times. d-Limonene is included in the list of Generally which enabled pharmacokinetic and pharmacodynamic correla-
Recognized As Safe (GRAS) agents listed by US FDA (FDA) and 10% tion (data not published). Rodents have a thinner stratum corneum
d-limonene was not found to cause any signicant pharmacological and higher hair follicles density than human skin, so it may overes-
effect (Fox et al., 2011). timate the permeability of drugs in human when using rodents skin
d-Limonene is known to have a potent skin permeation- as model. However, the recent research indicated that Sprague-
enhancing effect when used alone or in combination with PG Dawley rat is a useful model for predicting human skin permeability
(Williams and Barry, 1991). It can enhance both hydrophilic drugs with low inter-individual variations and similar permeation rate
including propranolol, sumatriptan succinate and lipophilic drugs (within twofold difference) (Takeuchi et al., 2011).
such as testosterone, ketoprofen, indomethacin and estradiol Considering the free-volatilized property of terpenes and
(Amnuaikit et al., 2005; Williams and Barry, 2004). A proposed ethanol which are not suitable for matrix type transdermal sys-
mechanism to improve the skin permeation of drugs is mainly the tem, a reservoir type transdermal delivery system was developed.
increase in drug diffusivity in the skin by modifying the intercellu- Reservoir-type TDS usually consists of a drug reservoir (as a solution
lar packing and disrupting highly ordered structure of lipids (Vaddi or gel) in between the impermeable backing layer and a rate con-
et al., 2002; Williams and Barry, 1991). These effects appear to trolling membrane. The rate controlling membrane can be either a
involve the modication of stratum corneum by solvent, improving microporous (e.g., polypropylene) or a nonporous polymeric mem-
drug partitioning into the tissue. Some authors have established brane (e.g., ethylene vinyl acetate copolymer with a specic drug
certain drug-specic structureactivity relationship for these ter- permeability). Pressure sensitive adhesive polymer was usually
penes (Drakulic et al., 2008; Ghafourian et al., 2004) based on the applied on the external surface of the polymer membrane to pro-
physicochemical properties and conformations of the complexes vide close contact of patch with skin surface (Babu and Pandit,
(drug + enhancer). According to this hypothesis, that hydrocarbon 2005). In the present study, in order to eliminate the inuence of the
or the non-polar group containing terpenes, such as limonene pro- pressure sensitive adhesive on the permeation of drug, the pressure
vide better enhancement for lipophilic permeates, and conversely, sensitive adhesive is coated on the peripheral region of the patch.
the polar group containing terpenes (such as menthol, 1,8-cineole) Transdermal patch of bufalin provided sustained release of
provide better enhancement for hydrophilic permeates (Williams bufalin and the MRT was 9.2 h, which is much longer than pre-
and Barry, 2004). In our experiments, d-limonene showed the high- viously developed lipid (2.4 h) and nano (7.6 h) formulations (Liu
est enhancing activity on the transdermal absorption of bufalin, et al., 2011; Yin et al., 2012). The optimization for future design of
which may be explained by the above mechanism. In order to better bufalin is to improve its low bioavailability (8%) through increas-
homogenize limonene in the aqueous matrix, we added 1% Tween ing the patch size or reduce of the dose. From deconvolution
80 to the formulation. We did not nd any signicant permeation analysis, both in vivo release rate and accumulative permeation
difference between the formulation with and w/o Tween 80. amounts of bufalin from transdermal patch indicated that bufalin
Ethanol is one of the most widely used solvents in many can maintain sustained release up to 12 h. The release rate of
transdermal formulations. The synergistic effect of ethanol with bufalin from batch slightly declined with time after 8 h, the pos-
d-limonene is better than PG can be interpreted by several sible reasons may due to decreased concentrations of ethanol and
mechanisms that have been proposed for ethanol permeation d-limonene in transdermal reservoir upon their fast permeation to
enhancing activity (Panchagnula et al., 2001). Firstly, it may skin. Loose contact of patch with skin surface in the later stage of
increase the solubility of bufalin in the vehicle compared with PK study due to movement of animal may also contribute to it.
Z. Yang et al. / International Journal of Pharmaceutics 447 (2013) 231240 239

In the current study, the highest level IVIVC (type A), which is Drakulic, B.J., Juranic, I.O., Eric, S., Zloh, M., 2008. Role of complexes formation
a point to point correlation of in vitro skin permeation to in vivo between drugs and penetration enhancers in transdermal delivery? Int. J. Pharm.
363, 4049.
plasma AUC was established. We realized the potential limitations FDA Part 182 SUBSTANCES GENERALLY RECOGNIZED AS SAFE. In: www.fda.gov/
caused by different animal models (mice skin in vitro and rats PK Food/../GenerallyRecognizedasSafeGRAS/default.htm
in vivo) used for IVIVC. However, our consistent IVIVC and decon- Fox, L., Gerber, M., Plessis, J., Hamman, J., 2011. Transdermal drug delivery enhance-
ment by compounds of natural origin. Molecules 16, 1050710540.
volution results further demonstrated that these two models have Gannu, R., Palem, C.R., Yamsani, V.V., Yamsani, S.K., Yamsani, M.R., 2010. Enhanced
good correlations for the release of bufalin. The good correlation bioavailability of lacidipine via microemulsion based transdermal gels: formu-
indicated that the permeation study using mice skin could differ- lation optimization, ex vivo and in vivo characterization. Int. J. Pharm. 388,
231241.
entiate the rate order of formulations and help with formulation
Ghafourian, T., Zandasrar, P., Hamishekar, H., Nokhodchi, A., 2004. The effect of pene-
screening and development. On the basis of in vitro permeation tration enhancers on drug delivery through skin: a QSAR study. J. Control. Release
results, Gannu et al. (2010) successfully established ex vivoin vivo 99, 113125.
Hashimoto, S., Jing, Y., Kawazoe, N., Masuda, Y., Nakajo, S., Yoshida, T., et al., 1997.
correlation to estimate systemic concentration of lacidipine fol-
Bufalin reduces the level of topoisomerase II in human leukemia cells and affects
lowing transdermal gels administration in rabbits. It also showed the cytotoxicity of anticancer drugs. Leuk. Res. 21, 875883.
similar biphasic prole between in vitro permeation across mouse Hong, S.H., Choi, Y.H., 2012. Bufalin induces apoptosis through activation of both the
skin and plasma. The possible reason for the initial fast absorption intrinsic and extrinsic pathways in human bladder cancer cells. Oncol. Rep. 27,
114120.
may due to fast transport and removal of bufalin by rich capillary of Hong, Z., Chan, K., Yeung, H.W., 1992. Simultaneous determination of bufadieno-
skin in live animal than excised skin. The real mechanism remains lides in the traditional Chinese medicine preparation, liu-shen-wan, by liquid
to be explored. After achieving steady state, in vitro permeation chromatography. J. Pharm. Pharmacol. 44, 10231026.
Kawazoe, N., Watabe, M., Masuda, Y., Nakajo, S., Nakaya, K., 1999. Tiam1 is involved
showed excellent IVIVC with correlation coefcient equals to 0.97. in the regulation of bufalin-induced apoptosis in human leukemia cells. Onco-
This excellent IVIVC relationship demonstrated that in vitro exper- gene 18, 24132421.
iments can be used to screen formulations and predict preclinical Krishnaiah, Y.S., Satyanarayana, V., Bhaskar, P., 2002. Effect of limonene on the
in vitro permeation of nicardipine hydrochloride across the excised rat abdom-
and clinical pharmacokinetics in further studies. inal skin. Pharmazie 57, 842847.
Li, D., Qu, X., Hou, K., Zhang, Y., Dong, Q., Teng, Y., et al., 2009. PI3K/Akt is involved in
bufalin-induced apoptosis in gastric cancer cells. Anticancer Drugs 20, 5964.
5. Conclusions Liu, D., Feng, J.F., 2008. Determination of apparent aqueous solubility and apparent
oilwater partition coefcients of three bufadienolides composition. Zhongguo
Clinical application of bufalin was mainly limited due to its nar- Zhong Yao Za Zhi 33, 12561258.
Liu, Y., Chen, Z.Q., Zhang, X., Feng, N.P., Zhao, J.H., Wu, S., et al., 2010. An improved
row therapeutical window and short half life. Transdermal drug formulation screening and optimization method applied to the development of
delivery of bufalin provides a sustained release rate and avoids a self-microemulsifying drug delivery system. Chem. Pharm. Bull. (Tokyo) 58,
the peak concentration, which prolong the duration of action and 1622.
Liu, Y., Wang, P., Sun, C., Zhao, J., Du, Y., Shi, F., et al., 2011. Bioadhesion and
decrease the risk of side effect. The present transdermal patch of enhanced bioavailability by wheat germ agglutinin-grafted lipid nanoparti-
bufalin maintained therapeutical plasma concentration as long as cles for oral delivery of poorly water-soluble drug bufalin. Int. J. Pharm. 419,
patch remained on the animal for at least 12 h. Deconvolution anal- 260265.
Meng, Z., Yang, P., Shen, Y., Bei, W., Zhang, Y., Ge, Y., et al., 2009. Pilot study of
ysis also proved its sustained release rate in vivo. IVIVC enabled huachansu in patients with hepatocellular carcinoma, nonsmall-cell lung can-
the prediction of pharmacokinetic prole of bufalin from in vitro cer, or pancreatic cancer. Cancer 115, 53095318.
permeation results. Our results demonstrated that transdermal Panchagnula, R., Salve, P.S., Thomas, N.S., Jain, A.K., Ramarao, P., 2001. Trans-
dermal delivery of naloxone: effect of water, propylene glycol, ethanol and
administration is an enabling formation strategy to deliver ade-
their binary combinations on permeation through rat skin. Int. J. Pharm. 219,
quate bufalin across skin on rodent models. Both in vitro and in vivo 95105.
studies showed consistent results that with optimized chemical Panesar, N.S., 1992. Bufalin and unidentied substance(s) in traditional Chi-
nese medicine cross-react in commercial digoxin assay. Clin. Chem. 38,
enhancer and formulation components, bufalin can achieve sus-
21552156.
tained and sufcient delivery across skin. Paudel, K.S., Milewski, M., Swadley, C.L., Brogden, N.K., Ghosh, P., Stinchcomb, A.L.,
2010. Challenges and opportunities in dermal/transdermal delivery. Ther. Deliv.
1, 109131.
Acknowledgements Qin, T.J., Zhao, X.H., Yun, J., Zhang, L.X., Ruan, Z.P., Pan, B.R., 2008. Efcacy and safety of
gemcitabineoxaliplatin combined with huachansu in patients with advanced
gallbladder carcinoma. World J. Gastroenterol. 14, 52105216.
This work was supported by research grants from Shanghai Sci- Song, H., Guo, T., Bi, K., Wang, H., Zhang, R., 2000. Determination of resibufogenin
ence Foundation (Shanghai, China). We thank Dr. Qinghua Ge and and cinobufagin in heart-protecting musk pill by HPLC. Biomed. Chromatogr.
her team for analytical assistance and Jin Lan for her value com- 14, 130132.
Sun, L., Cun, D., Yuan, B., Cui, H., Xi, H., Mu, L., et al., 2012. Formulation and in vitro/in
ments on this manuscript.
vivo correlation of a drug-in-adhesive transdermal patch containing azasetron.
J. Pharm. Sci. 101, 45404548.
Takeuchi, H., Mano, Y., Terasaka, S., Sakurai, T., Furuya, A., Urano, H., et al., 2011. Use-
Appendix A. Supplementary data fulness of rat skin as a substitute for human skin in the in vitro skin permeation
study. Exp. Anim. 60, 373384.
Supplementary data associated with this article can be Vaddi, H.K., Ho, P.C., Chan, Y.W., Chan, S.Y., 2002. Terpenes in ethanol: haloperidol
permeation and partition through human skin and stratum corneum changes?
found, in the online version, at http://dx.doi.org/10.1016/ J. Control. Release 81, 121133.
j.ijpharm.2013.02.048. Wang, L., Raju, U., Milas, L., Molkentine, D., Zhang, Z., Yang, P., et al., 2011. Huachansu,
containing cardiac glycosides, enhances radiosensitivity of human lung cancer
cells. Anticancer Res. 31, 21412148.
References Wang, S.W., Chiao, Y.C., Tsai, S.C., Lu, C.C., Chen, J.J., Shih, H.C., et al., 1997. Inhibition
of bufalin on pituitary and testicular function in rats. J. Pharmacol. Exp. Ther.
Amnuaikit, C., Ikeuchi, I., Ogawara, K., Higaki, K., Kimura, T., 2005. Skin permeation of 283, 528532.
propranolol from polymeric lm containing terpene enhancers for transdermal Williams, A.C., Barry, B.W., 2004. Penetration enhancers. Adv. Drug Deliv. Rev. 56,
use? Int. J. Pharm. 289, 167178. 603618.
Babu, R.J., Pandit, J.K., 2005. Effect of penetration enhancers on the release and skin Williams, A.C., Barry, B.W., 1991. Terpenes and the lipidprotein-partitioning theory
permeation of bupranolol from reservoir-type transdermal delivery systems. of skin penetration enhancement. Pharm. Res. 8, 1724.
Int. J. Pharm. 288, 325334. Yan, S., Qu, X., Xu, C., Zhu, Z., Zhang, L., Xu, L., et al., 2012. Down-regulation
Bozdogan, H., 2000. Akaikes information criterion and recent developments in infor- of Cbl-b by bufalin results in up-regulation of DR4/DR5 and sensitization of
mation complexity. J. Math. Psychol. 44, 6291. TRAIL-induced apoptosis in breast cancer cells. J. Cancer Res. Clin. Oncol. 138,
Dong, Y., Yin, S., Li, J., Jiang, C., Ye, M., Hu, H., 2011. Bufadienolide compounds sen- 12791289.
sitize human breast cancer cells to TRAIL-induced apoptosis via inhibition of Yang, Z., Luo, H., Wang, H., Hou, H., 2008. Preparative isolation of bufalin and cinob-
STAT3/Mcl-1 pathway. Apoptosis 16, 394403. ufagin from Chinese traditional medicine ChanSu. J. Chromatogr. Sci. 46, 8185.
240 Z. Yang et al. / International Journal of Pharmaceutics 447 (2013) 231240

Yang, Z., Zhu, W., Gao, S., Xu, H., Wu, B., Kulkarni, K., et al., 2010. Simul- Zhang, D.M., Liu, J.S., Tang, M.K., Yiu, A., Cao, H.H., Jiang, L., et al., 2012. Bufotalin from
taneous determination of genistein and its four phase II metabolites in Venenum Bufonis inhibits growth of multidrug resistant HepG2 cells through
blood by a sensitive and robust UPLCMS/MS method: application to an G(2)/M cell cycle arrest and apoptosis. Eur. J. Pharmacol. 692, 1928.
oral bioavailability study of genistein in mice. J. Pharm. Biomed. Anal. 53, Zhang, L., Nakaya, K., Yoshida, T., Kuroiwa, Y., 1992. Induction by bufalin
8189. of differentiation of human leukemia cells HL60, U937, and ML1 toward
Yeh, J.Y., Huang, W.J., Kan, S.F., Wang, P.S., 2003. Effects of bufalin and cinobufagin macrophage/monocyte-like cells and its potent synergistic effect on the differ-
on the proliferation of androgen dependent and independent prostate cancer entiation of human leukemia cells in combination with other inducers. Cancer
cells. Prostate 54, 112124. Res. 52, 46344641.
Yin, P., Wang, Y., Qiu, Y., Hou, L., Liu, X., Qin, J., et al., 2012. Bufalin-loaded Zhu, Z., Sun, H., Ma, G., Wang, Z., Li, E., Liu, Y., et al., 2012. Bufalin induces lung
mPEGPLGAPLLcRGD nanoparticles: preparation, cellular uptake, tissue dis- cancer cell apoptosis via the inhibition of PI3K/Akt pathway. Int. J. Mol. Sci. 13,
tribution, and anticancer activity. Int. J. Nanomed. 7, 39613969. 20252035.
Yu, C.H., Kan, S.F., Pu, H.F., Jea Chien, E., Wang, P.S., 2008. Apoptotic signaling Zorin, S., Kuylenstierna, F., Thulin, H., 1999. In vitro test of nicotines permeability
in bufalin- and cinobufagin-treated androgen-dependent and -independent through human skin. Risk evaluation and safety aspects. Ann. Occup. Hyg. 43,
human prostate cancer cells. Cancer Sci. 99, 24672476. 405413.

También podría gustarte