Zingiber cassumunar blended patches for skin application

Transcription

Zingiber cassumunar blended patches for skin application
a s i a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 1 0 ( 2 0 1 5 ) 3 4 1 e3 4 9
Available online at www.sciencedirect.com
H O S T E D BY
ScienceDirect
journal homepage: http://ees.elsevier.com/ajps/default.asp
Original Research Paper
Zingiber cassumunar blended patches for skin
application: Formulation, physicochemical
properties, and in vitro studies
Jirapornchai Suksaeree a,b,*, Laksana Charoenchai b, Fameera Madaka b,
Chaowalit Monton b, Apirak Sakunpak b,c, Tossaton Charoonratana b,c,
Wiwat Pichayakorn d,e
a
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Rangsit University, Muang, Pathum Thani 12000,
Thailand
b
Sino-Thai Traditional Medicine Research Center (Cooperation between Rangsit University, Harbin Institute of
Technology, and Heilongjiang University of Chinese Medicine), Faculty of Pharmacy, Rangsit University, Muang,
Pathum Thani 12000, Thailand
c
Department of Pharmacognosy, Faculty of Pharmacy, Rangsit University, Muang, Pathum Thani 12000, Thailand
d
Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University,
Hat-Yai, Songkhla 90112, Thailand
e
Medical Products Innovations from Polymers in Clinical Use Research Unit, Prince of Songkla University, Hat-Yai,
Songkhla 90112, Thailand
article info
abstract
Article history:
Our work was to study the preparation, physicochemical characterization, and in vitro
Received 15 December 2014
characteristic of Zingiber cassumunar blended patches. The Z. cassumunar blended patches
Received in revised form
incorporating Z. cassumunar Roxb. also known as Plai were prepared from chitosan and
16 February 2015
polyvinyl alcohol with glycerin as plasticizer. They were prepared by adding all ingredients
Accepted 3 March 2015
in a beaker and homogeneously mixing them. Then, they were transferred into Petri-dish
Available online 12 March 2015
and dried in hot air oven. The hydrophilic nature of the Z. cassumunar blended patches was
confirmed by the moisture uptake, swelling ratio, erosion, and porosity values. The FTIR,
Keywords:
DSC, XRD, and SEM studies showed revealed blended patches with amorphous region that
Chitosan
was homogeneously smooth and compact in both surface and cross section dimensions.
Polyvinyl alcohol
They exhibited controlled the release behavior of (E)-4-(30 ,40 -dimethoxyphenyl) but-3-en-l-
Z. cassumunar patches
ol (compound D) that is the main active compound in Z. cassumunar for anti-inflammation
Blended patches
activity. However, in in vitro skin permeation study, the compound D was accumulated in
Skin application
newborn pig skin more than in the receptor medium. Thus, the blended patches showed
the suitable entrapment and controlled release of compound D. Accordingly, we have
* Corresponding author. Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Rangsit University, Muang, Pathum Thani 12000,
Thailand. Tel.: þ66 (2) 9972222x1502, þ66 (2) 9972222x4911; fax: þ66 (2) 9972222x1403, þ66 (2) 9972222x1508.
E-mail address: [email protected] (J. Suksaeree).
Peer review under responsibility of Shenyang Pharmaceutical University.
http://dx.doi.org/10.1016/j.ajps.2015.03.001
1818-0876/© 2015 Shenyang Pharmaceutical University. Production and hosting by Elsevier B.V. This is an open access article under the
CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
342
a s i a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 1 0 ( 2 0 1 5 ) 3 4 1 e3 4 9
demonstrated that such chitosan and polyvinyl alcohol formulated patches might be
developed for medical use.
© 2015 Shenyang Pharmaceutical University. Production and hosting by Elsevier B.V. This is
an open access article under the CC BY-NC-ND license (http://creativecommons.org/
licenses/by-nc-nd/4.0/).
1.
Introduction
Topical and transdermal drug delivery systems are intended
for external use. They are often dermatologic products such as
sunscreens, local anesthetics, antiseptics and antiinflammatory agents intended for localized action on one or
more layers of the skin. Conversely, some transdermal drug
delivery systems are designed for percutaneous route of drug
delivery in which case skin is not the target. In such case, the
drug must be absorbed across the skin which is made up of
dermis and epidermis, especially the stratum corneum barrier
including sweat glands, sebaceous glands, and hair follicles
[1], and pass into deeper dermal layers to reach the systemic
blood circulation. Generally, substances intended for transdermal delivery systems are low molecular weight (100500 Da), potent non-irritation and non-allergenic [2e4]. The
delivery system can be categorized as either i) drug in adhesive or ii) drug in matrix systems. The drug is dispersed or
dissolved in a polymer matrix and attached to an adhesive
layer that contacts the skin. In some cases, the polymer matrix can act as the adhesive layer. Polymer matrix layers and/
or the added adhesive layer act as a control of the rate of delivery [5,6].
Thai traditional medicines (herbal medicines) are popular
for the treatment of various symptoms and diseases and to
promote good health. Although the Western modern medicines are increasingly popular, Thai traditional medicines are
still widely used especially among the rural Thais. Herbal
medicines may contain variations of active ingredients parts
of plants, other plant materials, or combinations that
included herbs, herbal materials, herbal preparations, and
finished herbal products. Zingiber cassumunar Roxb., also
known as Thai name “Plai”, is a medicinal plant widely
cultivated in Thailand and tropical Asia. It is frequently used
as an ingredient in marketed phytomedicines [7,8]. The
rhizome of Z. cassumunar Roxb. has an anti-inflammatory
activity. It has been the source of Thai traditional herbal
remedies and extracts for topical application to alleviate
inflammation [9e11]. The chemical composition of the
rhizome oils of Z. cassumunar Roxb. has been previously reported [7,10,12e16]. The major constituents of the crude oils
are terpinen-4-ol, a- and b-pinene, sabinene, myrcene, a- and
g-terpinene, limonene, terpinolene, sabmene, and monoterpenes [12,17]. (E)-4-(30 ,40 -dimethoxyphenyl) but-3-en-l-ol
(compound D) is the main active compound in Z. cassumunar
that exhibits anti-inflammatory [11,15,18], analgesic and
antipyrectic [11,15,16] activity in experimental models. It is
also used as topical treatment for sprains, contusions, joint
inflammations, muscular pain, abscesses, and similar
inflammation-related disorders [19e21]. Thus, this work
used the compound D as the marker compound for in vitro
study.
Herbal patches are adhesive patches that incorporate the
herbal medicines or extracted herb. When applied to the skin
the active compound is released at a constant rate. Such
patches are recommended for smoking cessation, herbal body
detox foot patch, relief of stress, to increase sexuality, as insect repellants, as male energizer, to improve sleep, to postpone menopause, for rheumatoid arthritis, as herbal plasters
patches, etc. [22].
The aim of the current study was to prepare a Z. cassumunar
containing product incorporating the crude Z. cassumunar oil
in blended patches that consisted of chitosan and polyvinyl
alcohol (PVA) polymer matrix combination using glycerin as
plasticizer. Similarly prepared blended patches without crude
Z. cassumunar oil served as control. The patches were evaluated with regard to the physicochemical properties as moisture uptake, swelling ratio, erosion, porosity, Fourier
transform infrared spectroscopy (FTIR), differential scanning
calorimetry (DSC), X-ray diffraction (XRD), scanning electron
microscope (SEM), and in vitro release and skin permeation
studies.
2.
Materials and methods
2.1.
Materials
The Z. cassumunar rhizome powder was purchased from
Charoensuk Osod, Thailand. The Z. cassumunar powder was
extracted in 95% ethanol and filtered through a 0.45 mm of
polyamide membrane to obtain crude Z. cassumunar oil. Chitosan (degree of deacetylation ¼ 85%, mesh size 30) was purchased from Seafresh Industry Public Co., Ltd, Thailand. PVA
and glycerin were purchased from SigmaeAldrich, USA. All
organic solvents were analytical grade obtained from Merck
KGaA, Germany.
2.2.
Analytical method
An Agilent 1260 Infinity system (Agilent Technologies, USA.)
was used for this experiment with detection at 260 nm, a
4.6 mm 250 mm diameter, 5 mm particle size C18 column
(ACE 5, DV12-7219, USA.), a flow rate of 1 ml/min, and injection volume of 10 mL. The mobile phase was a gradient elution
of 2% acetic acid in ultrapure water (A) and methanol (B) of 60
to 50% of A, 50 to 30% of A, 30 to 20% of A, 20 to 50% of A,
50 to 60% of A, and 60% of A for 0e5 min, 5e15 min,
15e25 min, 25e30 min, 30e32 min, and 32e40 min, respectively [23]. The HPLC validation method of compound D provided a limit of detection of 0.20 mg/ml, the limit of
a s i a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 1 0 ( 2 0 1 5 ) 3 4 1 e3 4 9
quantification of 0.80 mg/ml, good accuracy (95.38e104.76%),
precision (less than 2%CV), and linearity with good correlation coefficient (r2) > 0.9999 in the required concentration
range of 2e40 mg/ml. The separation method and validation
method of compound D from crude Z. cassumunar oil was
described in previous publication [23,24].
2.3.
Z. cassumunar blended patches preparation
The chitosan was dissolved in 1% acetic acid in distilled water
in concentration of 3.5%w/v. The PVA was dissolved in
distilled water in concentration of 20%w/v. The blank blended
patches were prepared by 2 g of 3.5%w/v chitosan were mixed
together with 5 g of 20%w/v of PVA, and homogeneously
mixed with 2 g of glycerin as plasticizer to obtain clear polymer blended solution. The Z. cassumunar blended patches
were prepared as 3 g of the crude Z. cassumunar oil completely
dissolved in absolute ethanol and continuously mixed in
polymer blend solution. They were transferred into Petri-dish
and dried in hot air oven at 70 ± 2 C for 5 h. Finally, they were
peeled from Petri-dish and kept in desiccator until used.
2.4.
Evaluation of blank and Z. cassumunar blended
patches
2.4.1.
SEM photography
The surface and cross section of blank blended patches and Z.
cassumunar blended patches were placed onto copper stub and
then coated with gold in a sputter coater. They were photographed under SEM equipment (model: Quanta 400, FEI, Czech
Republic) with high vacuum and high voltage of 20 kV condition, with Everhart Thornley detector (ETD).
2.4.5.
FTIR study
The FTIR study employed the Attenuated Total Reflectance e
FTIR (ATR-FTIR) technique for the chitosan film, PVA film,
crude Z. cassumunar oil, blank blended patches, and Z. cassumunar blended patches. They were scanned at a resolution of
4 cm1 with 16 scans over a wavenumber region of 400 e
4000 cm1. The FTIR spectrometer (model: Nicolet 6700,
DLaTGS detector, Thermo Scientific, USA.) was used to
determine IR transmission spectra and record the characteristic peaks.
2.4.3.
DSC study
A DSC instrument (model: DSC7, Perkin Elmer, USA) was used
to investigate the endothermic transition of the substances
that also confirmed the compatibility of each ingredient. The 1
e 10 mg of sample was weighted in DSC pan, hermetically
sealed, and run in the DSC instrument at the heating rate of
10 C/min under a liquid nitrogen atmosphere from 20 C to
350 C.
2.4.4.
XRD study
The XRD (model: X'Pert MPD, PHILIPS, Netherlands) was
employed to study the compatibility of the chitosan, PVA,
blank blended patches, and Z. cassumunar blended patches.
The generator operating voltage and current of X-ray source
were 40 kV and 45 mA, respectively, with an angular of 5 e 40
(2q), and a stepped angle of 0.02 (2q)/s.
Moisture uptake, swelling ratio, and erosion studies
For determination of moisture uptake, swelling ratio and
erosion, 1 cm 1 cm patch specimens were used. For moisture uptake determination, the patch specimens were
weighed for their initial value (W0), then moved to a stability
chamber (model: Climate Chamber ICH/ICH L, Memmert
GmbH þ Co. KG, Germany) which controlled the temperature
at 25 ± 2 C and 75% relative humidity environment. The
specimens were removed and weighed until constant (Wu).
The percentage of moisture uptake was calculated by Equation (1) [25]
Moisture uptake ¼
ðWu W0 Þ
W0
(1)
The swelling ratio and erosion study were also determined
by drying patch specimens at 60 ± 2 C overnight. Then, they
were weighed (W0) and immersed in 5 ml of distilled water
and moved to stability chamber (model: Climate Chamber
ICH/ICH L, Memmert GmbH þ Co. KG, Germany) which
controlled the temperature at 25 ± 2 C and 75% relative humidity environment for 48 h. After removal of excess water,
the hydrated patches were weighed (Ws). They were then
dried again at 60 ± 2 C overnight, and weighed again (Wd). The
percentage of swelling ratio and the percentage of erosion
were calculated by Equations (2) And (3), respectively.
%Swelling ratio ¼
%Erosion ¼
2.4.6.
2.4.2.
343
ðWs W0 Þ
100
W0
ðW0 Wd Þ
100
W0
(2)
(3)
Porosity determination
After the patch specimens were equilibrated in water, the
volume occupied by the water and the volume of the membrane in the wet state were determined. The porosity of patch
specimens was obtained by Equation (4).
%Porosity ¼
ðW1 W2 Þ
100
dwater
wlt
(4)
where W1 and W2 ¼ the weights of the membranes in the wet
and dry states (g), respectively, dwater ¼ the density of pure
water at 20 C, and w, l, t ¼ the width (cm), length (cm), and
thickness (cm) of the membrane in the wet state, respectively
[26,27].
2.5.
The determination of compound D in patches
The blended patches were cut into 2 cm 2 cm specimens
from different sites. Each Plai patch sample was soaked with
ethanol in 10 ml volumetric flask, and sonicated at 25 C for
30 min. Then, the solution was sampled for 0.5 ml and
transferred into 100 ml volumetric flask and adjusted to volume of 100 ml with ethanol. The solution was filtered through
a 0.45 mm filter and analyzed with HPLC method.
2.6.
In vitro release study of compound D
The modified Franz-type diffusion cell having effective diffusion area of 1.77 cm2 was used for in vitro release and skin
344
a s i a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 1 0 ( 2 0 1 5 ) 3 4 1 e3 4 9
permeation study of compound D from the Z. cassumunar
blended patches. The receptor medium was 12 ml of isotonic
phosphate buffer solution pH 7.4: ethanol ¼ 80:20, thermoregulated with a water jacket at 37 ± 0.5 C and stirred
constantly at 600 rpm with a magnetic stirrer. The crude Z.
cassumunar oil was applied on the cellulose membrane
(MWCO: 3500 Da, CelluSep® T4, Membrane Filtration Product,
Inc., USA) which was used as a barrier between the donor
compartment and the receptor compartment. The Z. cassumunar blended patch preparations were cut and placed
directly on the donor cells. The 1 ml of receptor solution was
withdrawn at 0, 0.5, 1, 2, 3, 4, 6, and 24 h intervals, and
immediately replaced with an equal volume of fresh receptor
medium. The compound D content in these samples was
determined by an HPLC method.
2.7.
In vitro skin permeation study of compound D
The in vitro skin permeation of the compound D from the Z.
cassumunar blended patches was also carried out using a
modified Franz-type diffusion cell [28], and pig skin with
hair removed was an applied partitioning membrane [29,30].
The newborn pigs of 1.4e1.8 kg weight that had died by
natural causes shortly after birth were freshly purchased
from a local pig farm in Chachoengsao Province, Thailand.
The full thickness of flank pig skin was excised, hair was
surgically removed, and the subcutaneous fat and other
extraneous tissues were trimmed with a scalpel, cleaned
with isotonic phosphate buffer solution pH 7.4, blotted dry,
wrapped with aluminum foil and stored frozen. Before
permeation experiments, this isolated skin was soaked
overnight in isotonic phosphate buffer solution pH 7.4, and
mounted on the modified Franz-type diffusion cell with the
stratum corneum facing upward on the donor compartment. The crude Z. cassumunar oil and Z. cassumunar
blended patches were laid onto the isolated skin in the same
way as for the release study. The receptor compartment was
12 ml of isotonic phosphate buffer solution pH 7.4:
ethanol ¼ 80:20 and stirred constantly at 600 rpm by a
magnetic stirrer, at a constant temperature of 37 ± 0.5 C. A
1 ml of the receptor solution was withdrawn at 0, 0.5, 1, 2, 3,
4, 6, and 24 h intervals and an equal volume of fresh receptor medium was immediately replaced. The compound D
content in these samples was determined by the HPLC
method.
All in vitro release and skin permeation studies were performed in triplicate and the means of all measurements
calculated. The results were presented in terms of cumulative
percentage release or skin permeation as a function of time
using the following formula:
Cumulative percentage release or skin permeation
¼
Dt
100
Dl
(5)
where Dt was the amount of compound D released or
permeated from the Z. cassumunar blended patches at time t
and Dl was the amount of compound D loaded into the Z.
cassumunar blended patches.
3.
Results and discussion
3.1.
Evaluation of blank and Z. cassumunar blended
patches
Generally, the Z. cassumunar rhizomes were of deep yellow
color possessing a strong camphoraceous smell, warm, spicy,
and bitter taste [12,17,31]. The extraction of the Z. cassumunar
rhizome powder yielded a clear, high viscosity, yellow-orange
crude Z. cassumunar oil. The solvent extraction of plant materials likely produced oleoresin, which contained not only the
volatile compounds but also waxes and color pigments [32]. In
addition, Sukatta et al. 2009 reported two pathways for Z.
cassumunar rhizome extraction-hydro distillation and hexane
extraction. They confirmed that hydro distillation produced
the yellowish, low viscosity crude Z. cassumunar oil, while the
crude Z. cassumunar oil from the hexane extraction was
yellow-orange in color and had high viscosity. Commonly, our
work could confirm from its appearance that crude Z. cassumunar oil was obtained. Therefore, when crude Z. cassumunar
oil was added in blank blended patches, it produced the dark
yellow patches referred to as Z. cassumunar blended patches.
The photographs of blank blended and Z. cassumunar blended
patches were shown in previous reports by our research group
[33].
The SEM technique was used to photograph the high resolution morphology of the surface and cross section of blank
blended patches and Z. cassumunar blended patches (Fig. 1).
The surface of blank blended patches was homogeneously
smooth and dense with no visual pores (Fig. 1A). The surface
of Z. cassumunar blended patches became rough and uneven
as a result of widely distributed conglomeration and aggregation in the matrix of Z. cassumunar blended patches (photographed by digital camera and presented in previous
publication [33]) (Fig. 1B).
Recorded spectra are shown in Fig. 2. For the chitosan film,
the absorption peaks of stretching vibrations of eOH groups
broadly overlapped the stretching vibration of NeH ranging
from 3750 to 3000 cm1. The broad stretching vibrations of
CeH bond were observed at 2920e2875 cm1. The bending
vibrations of methylene and methyl groups were also absorbed at 1375 cm1 and 1426 cm1, respectively. The spectrum
bands in the range of 1680e1480 cm1 were identified as vibrations of carbonyl bonds of the amide group and vibrations
of protonated amine group. The vibrations of CO group
occurred in the range from 1160 cm1e1000 cm1. In addition,
the spectrum band located at around 1150 cm1 related to
asymmetric vibrations of CO in the oxygen bridge resulting
from deacetylation of chitosan. The spectrum bands at
1080e1025 cm1 were attributed to eCO of the ring COH, COC,
and CH2OH. Finally, the small spectrum peak at ~890 cm1
corresponded to wagging of the saccharide structure of chitosan [34]. Furthermore, the spectrum of acetic acid were
found at 3050, 1720, and 1432 related toeOH bond in carboxylic acid, CeO bond, and CeO bond, respectively. In addition,
the PVA spectrum showed both OeH stretching and CeO
stretching at 3449 and 1637 cm1, respectively [35,36].
The chitosan film, blank blended, and Z. cassumunar
blended patches weighed 1.662, 8.747, and 8.821 mg,
a s i a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 1 0 ( 2 0 1 5 ) 3 4 1 e3 4 9
345
Fig. 1 e Surface (£500 (A), £1000 (B), and £1500 (C)) and cross section morphology (£1000 (D), £1500 (E), and £5000 (F)) of
blank blended patches (upper) and Z. cassumunar blended patches (bottom) under SEM technique.
respectively. They were run with DSC instrument to study the
thermal behavior. The thermogram of chitosan film, blank
blended, and Z. cassumunar blended patches showed an initial
broad peak at 70.33 C with 231.35 J/g of enthalpy of peak (DH),
99.34 C with 188.307 J/g of DH, and 92.37 C with 78.76 J/g of
DH, respectively, which was attributed to evaporation of
moisture and represented the required energy to vaporize
water present in their samples. Moreover, the degradation
DSC peak of chitosan film broadly occurred at 323.67 C with
127.30 J/g of DH. In addition, the blank blended patches and Z.
cassumunar blended patches revealed high broad endothermic
peaks at 257.00 C with 363.24 J/g of DH and 261.00 C with
606.41 J/g of DH, respectively. Although the observed endothermic peaks in blank blended patches and Z. cassumunar
blended patches were slightly changed, there were no new
exo- or endo-thermic peaks in any experimental ranges indicating compatibility of all ingredients (Fig. 3).
The XRD technique was used to identify and characterize
crystalline and amorphous form of chitosan film, PVA film,
blank blended patches, and Z. cassumunar blended patches
that had been studied in range of 5e40 (2q values) (Fig. 4). The
X-ray diffraction profile of chitosan film showed peaks at ~10
and ~23 (2q). The intensity result of PVA film was 19.69
representing their semi-crystalline characters because of the
strong intermolecular interaction between PVA chains
through intermolecular hydrogen bonding [37]. Thus, the
chitosan and PVA film exhibited the semi-crystalline characteristics, but the XRD patterns of blank blended patches and Z.
cassumunar blended patches had broad diffraction halo of
amorphous region.
From above experimentals, the FTIR, DSC and XRD results
showed that there were no chemical interactions between any
components in blank blended patches or Z. cassumunar
blended patches.
Limpongsa and Umprayn (2008) reported that moisture
uptake, swelling ratio, erosion, and porosity values play
important roles for the release behavior of active compound in
matrix type patches [38]. Thus, this research evaluated these
variables as show in Fig. 5. We found that the moisture uptake,
swelling ratio, erosion, and porosity of blank blended patches
were 28.85 ± 4.17, 21.01 ± 5.38, 2.39 ± 0.41, and 1.92 ± 0.22%,
respectively. When crude Z. cassumunar oil was added in blank
blended patches, the moisture uptake, swelling ratio, erosion,
and porosity of blank blended patches were 28.51 ± 0.78,
Fig. 2 e FTIR spectra of chitosan, PVA, blank blended
patches, and Z. cassumunar blended patches.
Fig. 3 e DSC thermograms of chitosan, blank blended
patches, and Z. cassumunar blended patches.
346
a s i a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 1 0 ( 2 0 1 5 ) 3 4 1 e3 4 9
mobility of chitosan and PVA increased, therefore, increasing
the hydrodynamic volume of the polymer compact.
3.2.
Fig. 4 e XRD patterns of chitosan, PVA, blank blended
patches, and Z. cassumunar blended patches.
20.93 ± 5.88, 2.42 ± 0.98, 1.86 ± 0.24%, respectively, which
were not significantly different from blank blended patches.
These results are due to the fact that hydrophilic parts of ingredients could be dissolved and eroded from the blended
patches. The chitosan and PVA could swell and immediately
had the hydrated blended patches contents. The chains
In vitro release study of compound D
In vitro release of the crude Z. cassumunar oil released compound D calculated as cumulative percentage release
90.43 ± 19.28% after 24 h (Fig. 6). The almost 100% release of
compound D in 24 h might be due to rapid diffusion in the
receptor medium as a fast, initial burst during the first 6 h.
The amount of compound D in the Z. cassumunar blended
patches was 2.19 ± 0.16 mg/cm2. When the Z. cassumunar
blended patches were studied in in vitro, the cumulative percentage release of compound D was 81.49 ± 10.92% after 24 h
(Fig. 6). The release behavior was similar to the compound D
release behavior from crude Z. cassumunar oil that had a fast
initial burst release during the first 6 h. This behavior was
likely due to the compound D on the surface of patches might
be rapid diffusion. However, the effect may be attributed to
the moisture uptake, swelling ratio, erosion, and porosity
whereby the patch could absorb the moisture, and create a
space and a large free volume within the blended patches that
enhanced compound D diffusion [38]. Moreover, Guo et al.
2011 reported enhanced drug diffusion with amorphous matrix type patches [39] which supports our results in in vitro
study. The in vitro release kinetics model of compound D
provided a better fit to first-order model than to the zero-order
and Higuchi's model (Fig. 6).
3.3.
In vitro skin permeation study of compound D
The in vitro skin permeation study was carried out in a modified
Franz-type diffusion cell using newborn pig skin as a partition
Fig. 5 e The moisture uptake, swelling ratio, erosion, and porosity values of blank blended patches and Z. cassumunar
blended patches.
a s i a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 1 0 ( 2 0 1 5 ) 3 4 1 e3 4 9
347
Fig. 6 e In vitro release of compound D content from crude Z. cassumunar oil and Z. cassumunar blended patches and in vitro
release kinetics of zero order model (A), first order model (B), and Higuchi's model (C).
Fig. 7 e In vitro skin permeation of compound D content from crude Z. cassumunar oil and Z. cassumunar blended patches
and in vitro skin permeation kinetics of zero order model (A), first order model (B), and Higuchi's model (C).
348
a s i a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 1 0 ( 2 0 1 5 ) 3 4 1 e3 4 9
membrane. The mean cumulative amount of compound D
permeated from crude Z. cassumunar oil and Z. cassumunar
blended patches were 38.55 ± 18.48% and 36.72 ± 11.29% after
24 h, respectively (Fig. 7). Although another publication reported that glycerine could enhance drug permeability [40,41],
the Z. cassumunar blended patches contained only a small
amount of glycerin as plasticizer which was unlikely to affect
drug permeation. Moreover, compound D was only slightly
detected in the receptor medium. Because of its structure,
compound D exhibits less hydrophilicity than hydrophobicity
[11,13,15]. The in vitro skin permeation kinetics model of compound D provided a better fit to a first-order model than zeroorder and Higuchi's model (Fig. 7).
Thus, the newborn pig skins were removed from modified
Franz-type diffusion cell apparatus. They were cut into small
pieces and homogenized, and then were extracted in absolute
ethanol. These solutions were analyzed for the remaining
compound D content by HPLC method. They contained
60.54 ± 39.55% and 46.77 ± 17.93% compound D content in
crude Z. cassumunar oil and Z. cassumunar blended patches,
respectively. Thus, the compound D was highly accumulated
in newborn pig skin layer minimum permeation into receptor
medium. However, the underlying mechanisms for this effect
was never reported and will be further studied.
4.
Conclusion
In the current work prepared the Z. cassumunar blended patches
made from chitosan and PVA polymer blends incorporating the
crude Z. cassumunar oil. The surface and cross section were
photographed for morphology study under SEM technique and
the physicochemical properties evaluated by FTIR, DSC, XRD,
moisture uptake, swelling ratio, erosion, and porosity. The results revealed compatible, homogeneous, smooth, and
compact blended ingredients. The blended patches could
absorb the moisture that resulted in swelling of blended
patches. They were eroded which increased the number of
porous channels homogenously to pass compound D from Z.
cassumunar blended patches. The blended patches provided a
controlled release and skin permeation of compound D when
studied by modified Franz-type diffusion cell apparatus. Thus,
the blended patches could be suitably used for herbal medicine
application.
Acknowledgment
The authors reported no declaration of interests. The authors
are thankful to the Faculty of Pharmacy and the Research
Institute of Rangsit University (Grant No.74/2555) for financial
supports.
references
[1] Adrian CW. Structure and function of human skin. In:
Adrian CW, editor. Transdermal and topical drug delivery.
Illinois: Pharmaceutical Press; 2003. p. 1e25.
[2] Ghosh TK, Abraham W, Jasti BR. Transdermal and topical
drug delivery systems. In: Jasti BR, Ghosh TK, editors. Theory
and practice of contemporary pharmaceutics. Florida: CRC
Press; 2004. p. 423e455.
[3] Ghosh TK, Pfister WR, Yum SI. The development of
transdermal and topical therapeutic systems. In: Ghosh TK,
Pfister WR, Yum SI, editors. Transdermal and topical drug
delivery systems. New York: Informa Healthcare; 1997. p. 7.
[4] Williams AC. Transdermal and Topical Drug Delivery. In:
Williams AC, editor. Transdermal and topical drug delivery:
from theory to clinical practice. London: Pharmaceutical
Press; 2003. p. 178e187.
[5] Pichayakorn W, Suksaeree J, Boonme P, et al. Deproteinized
natural rubber as membrane controlling layer in reservoir
type nicotine transdermal patches. Chem Eng Res Des
2012;91:520e529.
[6] Wokovich AM, Prodduturi S, Doub WH, et al. Transdermal
drug delivery system (TDDS) adhesion as a critical safety,
efficacy and quality attribute. Eur J Pharm Biopharm
2006;64:1e8.
[7] Han A-R, Kim M-S, Jeong YH, et al. Cyclooxygenase-2
inhibitory phenylbutenoids from the rhizomes of Zingiber
cassumunar. Chem Pharm Bull 2005;53:1466e1468.
[8] Mabberley DJ. The plant-book. 3rd ed. Cambridge: Cambridge
University Press; 2008.
[9] Janpim K, Sakkumduang W, Nualkaew S, et al. The 2nd
International Conference on Applied Science (ICAS) and The
3rd International Conference on Science and Technology for
Sustainable Development of the Greater Mekong Sub-region
(STGMS). Luang Prabang, Lao: Souphanouvong University;
2011. p. 604e607.
[10] Kaewchoothong A, Tewtrakul S, Panichayupakaranant P.
Inhibitory effect of phenylbutanoid-rich Zingiber cassumunar
extracts on nitric oxide production by murine macrophagelike RAW264.7 cells. Phytother Res 2012;26:1789e1792.
[11] Panthong A, Kanjanapothi D, Niwatananun V, et al. Antiinflammatory activity of compounds isolated from Zingiber
cassumunar. Planta Med 1990;56:655.
[12] Bordoloi AK, Sperkova J, Leclercq PA. Essential oils of Zingiber
cassumunar roxb. From Northeast India. J Essent Oil Res
1999;11:441e445.
[13] Masuda T, Jitoe A. Phenylbutenoid monomers from the
rhizomes of Zingiber cassumunar. Phytochem
1995;39:459e461.
[14] Jeenapongsa R, Yoovathaworn K, Sriwatanakul KM, et al.
Anti-inflammatory activity of (E)-1-(3,4-dimethoxyphenyl)
butadiene from Zingiber cassumunar Roxb. J Ethnopharmacol
2003;87:143e148.
[15] Panthong A, Kanjanapothi D, Niwatananant W, et al. Antiinflammatory activity of compound D {(E)-4-(30 ,40 dimethoxyphenyl)but-3-en-2-ol} isolated from Zingiber
cassumunar Roxb. Phytomedicine 1997;4:207e212.
[16] Ozaki Y, Kawahara N, Harada M. Anti-inflammatory effect of
Zingiber cassumunar Roxb. and its active principles. Chem
Pharm Bull (Tokyo) 1991;39:2353e2359.
[17] Bhuiyan MNI, Chowdhury JU, Begum J. Volatile constituents
of essential oils isolated from leaf and rhizome of Zingiber
cassumunar Roxb. Bangladesh J Pharmacol 2008;3:69e73.
[18] Kanjanapothi D, Soparat P, Panthong A, et al. A uterine
relaxant compound from Zingiber cassumunar. Planta Med
1987;53:329e332.
[19] Pithayanukul P, Tubprasert J, Wuthi-Udomlert M. In vitro
antimicrobial activity of Zingiber cassumunar (Plai) oil and a
5% Plai oil gel. Phytother Res 2007;21:164e169.
[20] Pongprayoon U, Soontornsaratune P, Jarikasem S, et al.
Topical antiinflammatory activity of the major lipophilic
constituents of the rhizome of Zingiber cassumunar. Part I: the
essential oil. Phytomedicine 1997;3:319e322.
a s i a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 1 0 ( 2 0 1 5 ) 3 4 1 e3 4 9
[21] Pongprayoon U, Tuchinda P, Claeson P, et al. Topical
antiinflammatory activity of the major lipophilic
constituents of the rhizome of Zingiber cassumunar. Part II:
hexane extractives. Phytomedicine 1997;3:323e326.
[22] Rathva SR, Patel NN, Shah V, et al. Herbal transdermal
patches: a review. Int J Drug Dis Herb Res 2012;2:397e402.
[23] Suksaeree J, Madaka F, Monton C, et al. Method validation of
(E)-4-(3',4'-dimethoxyphenyl)-but-3-en-1-ol in Zingiber
cassumunar Roxb. with different extraction techniques. Int J
Pharm Pharm Sci 2014;6:295e298.
[24] Suksaeree J, Charoenchai L, Pichayakorn W, et al. HPLC
method development and validation of (E)-4-(3,4dimethoxyphenyl)-but-3-en-1-ol in Zingiber cassumunar
Roxb. from Thai Herbal Compress ball. Int J Pharm Pharm Sci
Res 2013;3:115e117.
[25] Rajesh N, Siddaramaiah H, Gowda DV, et al. Formulation and
evaluation of biopolymer based transdermal drug delivery.
Int J Pharm Pharm Sci 2010;2:142e147.
[26] Chen Z, Deng M, Chen Y, et al. Preparation and performance
of cellulose acetate/polyethyleneimine blend microfiltration
membranes and their applications. J Membr Sci
2004;235:73e86.
[27] Suksaeree J, Boonme P, Taweepreda W, et al. Relationships
between hydraulic permeability and porosity of natural
rubber blended films. Isan J Pharm Sci 2012;8:89e95.
[28] Venter JP, Müller DG, du Plessis J, et al. A comparative study
of an in situ adapted diffusion cell and an in vitro Franz
diffusion cell method for transdermal absorption of
doxylamine. Eur J Pharm Sci 2001;13:169e177.
[29] Meyer W, Schwarz R, Neurand K. The skin of domestic
mammals as a model for the human skin with special reference
to the domestic pig. Curr Probl Dermatol 1978;7:39e52.
[30] Simon GA, Maibach HI. The pig as an experimental animal
model of percutaneous permeation in man: qualitative and
quantitative observations e an overview. Skin Pharmacol
Appl Skin Physiol 2000;13:229e234.
[31] Sukatta U, Rugthaworn P, Punjee P, et al. Chemical
composition and physical properties of oil from plai (Zingiber
[32]
[33]
[34]
[35]
[36]
[37]
[38]
[39]
[40]
[41]
349
Cassumunar Roxb.) obtained by hydro distillation and hexane
extraction. Kasetsart J Nat Sci 2009;43:212e217.
Ibrahim J. Workshop on the extraction of essential oils.
Kepong, Malaysia: FRIM; 1997.
Suksaeree J, Monton C, Sakunpak A, et al. Physicochemical
properties study of Plai patches for topical applications. Int J
Pharm Pharm Sci 2014;6:434e436.
Silva SMLB, Carla RC, Fook MVL, et al. Application of infrared
spectroscopy to analysis of chitosan/clay nanocomposites.
In: Theophanides T, editor. Infrared spectroscopy e
materials science, engineering and technology. Croat:
InTech; 2012. p. 43e62.
Chhatri A, Bajpai J, Bajpai AK, et al. Cryogenic fabrication of
savlon loaded macroporous blends of alginate and polyvinyl
alcohol (PVA). Swelling, deswelling and antibacterial
behaviors. Carbohydr Polym 2011;83:876e882.
Kumar HMPN, Prabhakar MN, Prasad CV, et al. Compatibility
studies of chitosan/PVA blend in 2% aqueous acetic acid
solution at 30 C. Carbohydr Polym 2010;82:251e255.
Abdelaziz M, Ghannam MM. Influence of titanium chloride
addition on the optical and dielectric properties of PVA films.
Phys B 2010;405:958e964.
Limpongsa E, Umprayn K. Preparation and evaluation of
diltiazem hydrochloride diffusion-controlled transdermal
delivery system. AAPS PharmSciTech 2008;9:464e470.
Guo R, Du X, Zhang R, et al. Bioadhesive film formed from a
novel organiceinorganic hybrid gel for transdermal drug
delivery system. Eur J Pharm Biopharm 2011;79:574e583.
Pichayakorn W, Suksaeree J, Boonme P, et al. Deproteinized
natural rubber latex/hydroxypropylmethyl cellulose
blending polymers for nicotine matrix films. Ind Eng Chem
Res 2012;51:8442e8452.
Pichayakorn W, Suksaeree J, Boonme P, et al. Nicotine
transdermal patches using polymeric natural rubber as the
matrix controlling system: effect of polymer and plasticizer
blends. J Membr Sci 2012;411e412:81e90.