Anda di halaman 1dari 11

International Journal of Biological Macromolecules 101 (2017) 569–579

Contents lists available at ScienceDirect

International Journal of Biological Macromolecules


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

Development of Curcumin loaded chitosan polymer based


nanoemulsion gel: In vitro, ex vivo evaluation and in vivo wound
healing studies
Lydia Thomas a , Foziyah Zakir a , Mohd. Aamir Mirza b , Md. Khalid Anwer c ,
Farhan Jalees Ahmad a , Zeenat Iqbal a,∗
a
Nanomedicine Lab, Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi 110062, India
b
New Zealand Fulvic Limited, Mount Maunganui, Tauranga 3116, New Zealand
c
Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia

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

Article history: In the present study, various nanoemulsions were prepared using Labrafac PG + Triacetin as oil, Tween
Received 11 December 2016 80 as a surfactant and polyethylene glycol (PEG 400) as a co-surfactant. The developed nanoemul-
Received in revised form 10 March 2017 sions (NE1-NE5) were evaluated for physicochemical characterizations and ex-vivo for skin permeation
Accepted 13 March 2017
and deposition studies. The highest skin deposition was observed for NE2 with 46.07% deposition
Available online 16 March 2017
amongst all developed nanoemulsions (NE1-NE5). Optimized nanoemulsion (NE2) had vesicle size of
84.032 ± 0.023 nm, viscosity 78.23 ± 22.2 cps, refractive index 1.404. Nanoemulsion gel were developed
Keywords:
by incorporation of optimized nanoemulsion (NE2) into 1–3% chitosan and characterized by physical
Curcumin
Nanoemulsion
evaluation and rheological studies. Chitosan gel (2%) was found to be suitable for gelation of nanoemul-
Gel sion based on its consistency, feel and ease of spreadability. The flux of nanoemulsion gel was found
68.88 ␮g/cm2 /h as compared to NE2 (76.05 ␮g/cm2 /h) is significantly lower suggesting limited skin per-
meation of curcumin form gel. However, the retained amount of curcumin on skin by gel formulation
(980.75 ± 88 ␮g) is significantly higher than NE2 (771.25 ± 67 ␮g). Enhanced skin permeation of NE2
(46.07%) was observed when compared to nanoemulsion gel (31.25%) and plain gel (11.47%). The out-
come of this study evidently points out the potential of curcumin entrapped nanoemulsion gel in wound
healing.
© 2017 Elsevier B.V. All rights reserved.

1. Introduction of serious side effects or the disadvantage of early inactivation of


the drug.
Healing wound is of perennial interest and fundamental impor- Studies on curcumin revealed its antioxidant activity that may
tance to the surgeon. Wound is typically a tissue injury which have play an excellent role in the management of wound healing
involves inflammation followed by oxidative stress mediated by the [3]. Curcumin, a natural product obtained from the rhizomes of
formation of free radicals thus leading to DNA breakage, lipid per- Curcuma longa has been reported to be effective in wound repair
oxidation and enzymes inactivation. Various factors such as such as used both orally and topically. Curcumin is a o-methoxyphenol
diabetes, infection or metabolic deficiencies are susceptible to fail- derivative which possesses antioxidant activity and stimulates
ure in healing of wound [1]. A variety of therapeutic regimes have detoxification enzymes. It has been reported that when curcumin is
been investigated experimental and clinical settings for accelera- applied topically it significantly improves wound healing and pre-
tion of wound healing [2]. However, treatment of wounds by oral vents oxidative damage [4]. Further curcumin treatment result in
and parenteral application of drugs can be accompanied by the risk an increased formation of granulation in tissue, including greater
cellular content and neo vascularisation and a faster re epithelial-
isation of the wound [5]. However being a poorly water soluble
drug it is preferably localized in the superficial stratum corneum
(SC) after topical administration. Nevertheless using an optimal
∗ Corresponding author at: Dept. of Pharmaceutics, Jamia Hamdard, New Delhi vehicle, even for lipophilic substances like curcumin, it is possi-
110062, India. ble to minimize this accumulation. In addition, direct application
E-mail address: ziqbaljh@yahoo.co.in (Z. Iqbal).

http://dx.doi.org/10.1016/j.ijbiomac.2017.03.066
0141-8130/© 2017 Elsevier B.V. All rights reserved.
570 L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579

of curcumin to the skin is a matter of concern because of toxic [15]. To construct pseudoternary phase diagrams, the selected
effects of high concentration of polyphenols. Therefore a formu- oil phase (Labrafac PG: Triacetin, 1:1) was mixed with surfactant:
lation which releases the drug slowly in a controlled manner will cosurfactant mixture (Tween 80 and PEG 400, Smix ). The ratios of
be acceptable for efficient delivery of curcumin. Hence a water Smix used for titration are 1:0, 1:1, 1:2, 1:3, 1:4, 2:1, 3:1 and 4:1
based controlled released formulation such as curcumin loaded and the mixture was titrated with distilled water until it turned
nanoemulsion (NE) can be a suitable approach for cutaneous wound turbid. The volume of water used was recorded. Water titration
healing [6]. Recently, a number of investigations have been car- was continued until a clear, isotropic and thermodynamically sta-
ried out which demonstrated that drugs incorporated into NEs ble dispersion with low viscosity was obtained. The pseudo ternary
penetrate efficiently into the skin and through the subcutaneous phase diagrams were constructed by plotting water phase, oil
barrier [7].So it is imperative to design such a formulation which phase and surfactant: cosurfactant phase (Smix ) used in the exper-
enhances the solubilisation of curcumin and improves its perme- iment. Different formulations were selected from pseudoternary
ation through the skin. Many literatures are available regarding phase diagrams for thermodynamic evaluation. The best ratio was
enhancement of solubility and dissolution by different techniques selected for the development of curcumin loaded NEs [16].
such as commplexation, nanoemulsion, polymeric nanoparticles
etc [8,9]. 2.4. Thermodynamic stability of nanoemulsions (NEs)
The encapsulation of active ingredients into nanoemulsions as
carrier systems is a novel approach for drug delivery. NEs are In order to select the stable NEs and to discard the unstable
defined as clear, thermodynamically stable, isotropic mixture of or metastable ones, the drug loaded NEs were subjected to ther-
oil, water and surfactant/cosurfactant combination [10]. Lipophilic modynamic stability testing, which comprises of heating cooling
drugs usually get localised in the superficial skin layers after topical cycles. The formulations were kept between 37 ◦ C and 4 ◦ C for
application. Recent investigations show lipophilic drugs incorpo- 24 h (3 cycles) at each temperature and observed for any physi-
rated into NEs penetrate efficiently into the skin. As topical vehicles, cal changes. Various aspects like phase separation, turbidity etc. at
NEs can increase the local or systemic delivery of a drug by differ- room temperature were observed. The selected stable formulations
ent mechanisms [11]. First their composition and structure enable were subjected to centrifugation for 30 min at 5000 rpm. The stable
them to incorporate a greater amount of drug than other conven- formulations which did not show any phase separation or turbidity
tional topical formulations like ointments, creams, gels and lotions. were treated for freeze thaw cycles at −20 ◦ C for 24 h and −20 ◦ C for
NEs can enhance the solubility of poorly water-soluble drugs via 2–3 min (3 cycles). The NEs which returned to their original form
the finely dispersed oil droplet phase. Second the diffusional bar- within 2–3 min were selected for further studies [17].
rier of the skin may be modified depending on the composition
of the NE [12]. Third an increased thermodynamic activity of the 2.5. Nanoemulsion characterisation
drug may favour its partitioning into the skin [13]. Thus the formu-
lation will enable the drug to reach the underlying skin layers of 2.5.1. Transmission electron microscopy
the wound, specifically to the stratum germinativum and the der- Morphology of the NE was studied using TEM (Morgagni 268D
mis where wound repair and angiogenesis take place and act as a SEI, USA) operating at 200 kV and a 0.18 nm capable of point to
microreservoir with a slow release of the entrapped agent. point resolution. Combination of bright field imaging at increasing
magnification and diffraction modes was used to reveal the form
2. Materials and methods and size of the NE. In order to perform the TEM observations, the
diluted NE was deposited on the holey film grid and observed after
2.1. Materials drying [18].

Curcumin was obtained from Sigma Aldrich, (France). Triacetin 2.5.2. Measurement of droplet size
was obtained from Central Drug House, (Mumbai). Labrafac PG was Droplet size was determined by photon correlation spec-
a gift sample from Gattefosse (Saint Priest, France). Tween 80 and troscopy that analyzes the fluctuations in light scattering due to
Polyethylene glycol (PEG 400) were purchased from (Schuchardh, brownian motion of the particles [19] using a Zetasizer (1000 HS,
Germany). All others chemicals were of analytical grade. Malvern Instruments, UK). The formulation (0.1 mL) was dispersed
in 10 ml of water in a volumetric flask, mixed thoroughly with vig-
2.2. Determination of the solubility of curcumin in oils and orous shaking and light scattering was monitored at 25 ◦ C at a 90◦
surfactants angle.

The solubility studies were carried out as described by Yuan 2.5.3. Viscosity
et al., 2008 [14], with slight modification. Briefly, 2 ml of different The viscosity of the formulations was determined using Brook-
oils were taken in small vials and excess amount of the curcumin field DV III ultra V6.0 RV cone and plate rheometer (Brookfield
was added. The vials were tightly stoppered and continuously Engineering Laboratories, Inc., Middleboro, MA) using spindle #
stirred for 72 h in mechanical shaker at 37 ± 5 ◦ C and the samples CPE40 at 25 ± 0.5 ◦ C [18].
were centrifuged at 10,000 rpm for 10 min. The supernatant was
separated, filtered and appropriately diluted with methanol and 2.5.4. Refractive index and pH
the amount of drug content was quantified by spectrophotometric The refractive index of the NEs were determined using an Abbes
method. The same method was followed for determination of solu- type refractometer (Nirmal International, New Delhi) at 25 ± 5 ◦ C.
bility of curcumin in surfactants and cosurfactants. The selected oils The apparent pH of the formulation was measured by pH meter
and surfactants were further used for the construction of pseudo- (Accument AB15, Fischer Scientific, USA) in triplicate at 25 ± 1 ◦ C
ternary phase digrams as discussed below. [18].

2.3. Construction of pseudo-ternary phase diagrams 2.6. Formulation of optimized nanoemulsion gel

For the determination of existence zone of NE, pseudoternary The optimised NE was incorporated into gel by using chitosan
phase diagrams were constructed using aqueous titration method 2% as gelling agent. Chitosan was mixed with 1% aqueous solution
L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579 571

of acetic acid manually in a mortar. After that it was hydrated for 1 h 2.10. Permeation data analysis
at room temperature to obtain a gel of desirable viscosity and com-
patibility with the nanoemulsions [20]. 0.025% cetyl pyridinium The cumulative amount of curcumin permeated through the
chloride was incorporated into the chitosan gel as an antiseptic. skin (Q, ␮g/cm2 ) was plotted as a function of time (hr). The drug
The optimized NE was then mixed with the gel in 1:1 ratio with flux (permeation rate) at steady state (Js, ␮g/cm2 /h) was calculated
gentle stirring. from the slope of linear portion of the curve [26].

2.7. Evaluation of nanoemulsion gel formulation Cumulative amount of drug permeated

2.7.1. Drug content Concentration (g/ml)x Volume of diffusion cell


=
For determination of drug content, about 1 g of nanoemulsion Area (cm2 )
gel (NEG) formulation was weighed in a 100 ml volumetric flask and
dissolved in methanol. It was diluted appropriately and analyzed
at 420 nm by UV spectrophotometry. Permeability coefficient (Pb )
Flux
=
2.7.2. Viscosity and pH Drug concentration in donor compartment (g/ml)
The viscosity and pH of the NEG formulation was determined
using methods described previously [18].

2.11. Skin deposition study


2.7.3. Spreadability
Spreadbility of the NEG is an important parameter from patient For optimum wound healing it is necessary that a reasonable
compliance point of view. Briefly, 0.5 g test formulation was placed amount of the drug be retained in the skin. Skin deposition stud-
within a circle of 1 cm diameter pre-marked on a glass plate over ies were conducted in order to quantify the amount of curcumin
which a second glass plate was added [21]. A weight of 500 g was deposited in the skin. After 24 h of ex-vivo permeation study, the
allowed to rest on the upper glass plate for 5 min. The increase in skin surface was washed five times with ethanol:water (1:1), fol-
the diameter due to spreading of the test formulation was noted lowed by water to remove excess drug from the surface. The skin
[22]. was then cut in to small pieces and homogenized with methanol
(5 ml) by Teflon homogenizer for 6 h at room temperature. After
shaking for 5 min, it was centrifuged for 5 min at 5000 rpm and
2.8. Preparation of rat skin
supernatant was collected, filtered using 0.45 ␮m membrane fil-
ter and analyzed for curcumin content by UV spectrophotometer
Albino rats (200–250 g) obtained from central animal facility at
at ␭max 420 nm [27]. The skin permeation and deposition studies
Jamia Hamdard (protocol number 526) were sacrificed with pro-
were carried out for optimised nanoemulsion gel formulation in a
longed ether anesthesia. Full thickness rat skin was excised from
similar manner.
the abdomen and hair on the skin was removed with depilatory
cream. Subcutaneous tissue was surgically removed and dermis
side was wiped with isopropyl alcohol to remove residual adhering 2.12. Skin permeation and deposition studies
fat. The skin was washed with distilled water and stored in deep
freezer at −20 ◦ C till further use [23]. Skin permeation and deposition studies for optimized NEG were
carried out according to the procedure as described previously.
The NEG formulation permeation parameters were also compared
2.9. Skin permeation study
with plain curcumin gel (control) in order to elucidate the effect of
nanoemulsion on skin permeation of curcumin.
Ex-vivo permeation studies through rat skin was performed
using an automated diffusion cell sampling system (SFDC6, LOGAN
Inst, NJ, USA). Following pre-treatment, the skin was cut and 2.13. Skin irritation test
trimmed to appropriate size and mounted on a vertical diffusion
sampling cell. Epidermal membrane samples were mounted into The animal protocol (number 526) to carry out skin irritation
the diffusion cells (area 0.653 cm2 ) equilibrated at 37 ± 0.5 ◦ C for study was reviewed and approved by Institutional Animal Ethics
8–10 h. The stratum corneum of the skin faced the drug donor Committee, Jamia Hamdard and their guidelines were followed
cell whereas dermal side was in contact with the receptor phase. for the studies. The skin irritation test of the optimised NEG for-
The donor and receptor compartment was filled to capacity with mulation was carried out on Swiss albino mice weighing 20–25 g.
acetate buffer pH 5.5 [24]. The receiver fluid was stirred by a Teflon- The animals were kept under standard laboratory conditions, with
coated mini magnetic bar at a speed of 600 rpm and equilibrated at temperature of 25 ◦ C ± 1◦ C and relative humidity of 55% ± 5%. The
37 ± 0.5◦ C for 8–10 h. After application of the test formulation on animals were housed in polypropylene cages, 6 per cage, with free
the donor side, 0.5 ml of aliquot was collected from the receiver access to a standard laboratory diet and water ad libitum. The ani-
cell at designated time intervals (viz. 0, 1, 2, 3,4, 5, 6, 8, 10, 12 mals were divided into 3 groups as follows:
and 24 h) for 24 h period and replaced immediately with the same Group I Control (Plain curcumin gel)
volume of fresh media maintained at 37± 0.5◦ C. After appropriate Group II Blank Nanoemulsion gel (Nanoemulsion gel without
dilutions, the samples were filtered using 0.45 ␮m membrane filter curcumin)
and the amount of drug in the receptor media was analyzed using Group III Curcumin loaded nanoemulsion gel (NEG)
UV spectrophotometer at 420 nm. Receiver volume was immedi- A single dose of the NEG containing 2.5 mg curcumin was
ately replenished with the same amount of fresh acetate buffer pH applied to the left ear of the mouse, with the right ear as control.
5.5. Ethanol was added to the buffer to increase the solubility of The development of erythema was monitored for 6 days using a
curcumin for maintaining sink conditions [25]. method established by Van- Abbe et al. [28].
572 L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579

70
60
Solubility (mg/ml)
50
40
30
20
10
0

OILS SURFACTANTS CO-SURFACTANTS

Fig. 1. Solubility of curcumin in various oils, surfactants and co-surfactants (n = 3).

2.14. Stability studies 3. Result and discussion

The optimised NEG was stored at different temperatures for 3 3.1. Solubility of curcumin
months. As per ICH guidelines, it was stored at 25 ± 2 ◦ C and 75 ± 5%
RH. Then the consistency and concentration of curcumin was inves- The solubility studies of curcumin were carried out in selected
tigated at the end of 0, 30, 60, 90 days. The samples were withdrawn, oils, surfactants and co-surfactants and their results are depicted in
diluted with methanol and analysed using UV spectrophotometer. Fig. 1. Higher solubilities were found in synthetic oils as compared
The amount of drug degraded and the shelf-life of the formulation to edible natural oils. The results are in accordance to previous
was determined from Arrhenius plot. reports that edible oils are incapable of dissolving large amounts
of lipophillic drugs. The maximum solubility of curcumin was
found in Sefsol, Labrafac PG and Triacetin with solubility values of
2.15. In-vivo wound healing study 14 ± 2.1 mg/ml, 17.5 ± 0.9 mg/ml and 20 ± 1.1 mg/ml respectively.
However because of insignificant differences in curcumin solu-
The animal protocol (number 526) to carry out in vivo study was bility in these three oils, their combination was also evaluated
reviewed and approved by Institutional Animal Ethics Committee, for solubility. Among the various combinations, namely Sefsol:
Jamia Hamdard and their guidelines were followed for the stud- Labrafac (1:1), Sefsol: Triacetin (1:1), Labrafac PG: Triacetin (1:1)
ies. Albino rats, 7–9 weeks old and weighing 200–250 g were used and Labrafac PG: Triacetin (1:2), the drug exhibited highest solu-
for the study. Screening for wound-healing activity was performed bility (25 ± 1.2 mg/ml) in Labrafac PG, Triacetin mixture in a ratio
by incision wound model. The hair on the skin of back surface of of 1:1. Since curcumin is hydrophobic, we aim to formulate o/w
animals was removed by using a suitable depilatory cream (Anne nanoemulsion. Hence the amount of drug incorporated in oil phase
French hair removing cream). The animals were starved for 12 h is an important criterion as poor soluble drugs needs more amounts
prior to wounding. After shaving the backs of the animals, a lin- of oil for maximum drug loading this will increase the cost of
ear 3 cm incision was made over the skin of the back. Eighteen the formulation. Further, such a formulation will demand higher
animals were randomly divided into three groups namely, experi- amounts of surfactant blends to produce a stable NE [30].
mental control, placebo and treatment groups. The vehicle control The choice of surfactant is another important factor as the right
group did not receive any treatment. Placebo group received the blend with sufficient HLB value can form stable nanoemulsion. In
NEG devoid of curcumin and the treatment group received NEG order to formulate o/w NE, the HLB value of surfactant mixture
with curcumin. Application was carried out once in a day. For com- should be more than 10. To achieve this purpose, we tried vari-
putation of the percentage of wound healing, a transparent paper ous non-ionic surfactants such as Tween 80, Tween 20, Labrafil and
was placed on the location of wound and its shape was drawn on the Labrasol due its least toxicity as compared to their counterparts.
paper. Then the wound area was measured by matching the shape It is imperative that we conduct solubility study of curcumin in
of wound with a graph paper. The percentage of wound healing was surfactant and co-surfactant so that the surfactant with highest sol-
calculated by walker formula after measurement of the wound area. ubility can be selected. Due to toxic nature of surfactants, they can
Percentage of wound healing was computed every three days upto only be used in limited amounts thus necessitating their judicious
the 18th day [29]. selection. Curcumin was found to be highly soluble in Tween 80
(60 ± 3.1 mg/ml) when compared with other surfactants. Therefore,
Tween 80 was selected for our study. Generally short to medium
Wound area on day X
Percentage of wound area = chain alcohols are used as co-surfactants. The solubility of cur-
Wound area on day 1
cumin in various co-surfactants was also carried out and maximum
solubility was found in PEG 400 (45 ± 3.1 mg/ml). An important cri-
teria which cannot be overlooked is the safety profile of surfactants
Percentage of wound healing = 100 − Percentage of wound area and co-surfactants [16]. All the surfactants selected are pharma-
ceutically acceptable and categorized as generally regarded as safe
L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579 573

WATER WATER WATER

A Smix: 1:0 B C
Smix 1:1 Smix 1:2

OIL Smix OIL Smix OIL Smix

WATER WATER
WATER

D E F
Smix 1:3 Smix 1:4 Smix 2:1

OIL Smix OIL Smix OIL Smix

WATER WATER

G H
Smix 3:1 Smix 4:1

OIL Smix OIL Smix

Fig. 2. Pseudoternary phase diagrams indicating oil-in-water nanoemulsion (shaded area) region of Labrafac PG and Triacetin (oil), Tween 80 (surfactant), and PEG 400
(cosurfactant) at different Smix ratios; A (Smix 1:0), B (Smix1:1), C (Smix1:2), D (Smix 1:3), E (Smix 1:4), F (Smix 2:1), G (Smix 3:1), H(Smix 4:1).
Yellow dots – nanoemulsion zone; Purpule dots – microemulsion zone.

“GRAS” but only within a limited quantity. Therefore using a blend 3.2. Pseudo-ternary phase diagram
of surfactant and co-surfactant allows the use of low concentrations
of individual surfactant, which is a suitable approach to reduce the The pseudo-ternary phase diagrams were constructed to elu-
toxicity of the formulation (development of o/w). Additionally, HLB cidate any changes in the area of NE region with change in
value of the selected surfactant blend i.e. Tween 80 and PEG 400 was excipients. The Figures show phase diagrams constructed using
found to be 13.4, which fulfills the criteria of minimum HLB value Labrafac PG:Triacetin (1:1) as oil phase and different Smix ratios
required for the formulation of a stable o/w NE. of Tween 80 and PEG 400; 1:0, 1:1, 1:2, 1:3, 1:4, 2:1, 3:1, 4:1.
574 L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579

The relationship between the phase behaviour of a mixture and its Table 1
Compositions oil, Smix and water for selected nanoemulsion formulations.
composition can be captured with the aid of a phase diagram. Pseu-
doternary phase diagrams were constructed separately for each Nanoemulsions % v/v components S/Cos ratio
Smix ratio (Fig. 2), so that o/w NE regions could be identified and
Oil Smix Distilled water
NE formulations could be optimized. The translucent nanoemulsion
NE1 12 40 48 1:1
region is presented in phase diagrams. The rest of the region on the
NE2 15 40 45 2:1
phase diagram represents the turbid and conventional emulsions NE3 18 40 42 2:1
based on visual observation. In Fig. 2A, B and H), Two color could NE4 15 35 50 3:1
be seen (Yellow and Purpule). During phase diagram construction NE5 18 36 46 4:1
two zone were selected in software. Yellow and purpule color in
phase diagram describes nanoemulsion and microemulsion zone
respectively. It is clearly evident from the graph (Fig. 2A), NE pre-
pared without any co-surfactant exhibit a low NE region. However
when the co-surfactant is added this region has increased signif- drug can be solubilised per ml of oil. Therefore 12% was selected as
icantly. Further when the Smix ratio was increased from 1:1 to the least oil concentration to be taken for 1 ml of formulation from
1:3, the nanoemulsion area also increased, though insignificantly the phase diagram. Curcumin when applied topically in a concen-
(Fig. 2B–D). From these observations, it can be inferred that the tration of 0.1% has been found to be effective for wound healing [5].
use of a single surfactants cannot appreciably reduce the interfa- Keeping this in mind the drug stock solutions were prepared in such
cial energy required for the formation of NE. Therefore a second a way that 2.5 mg dose is present in each formulation complying the
surfactant known as co-surfactant is generally added, which will oil percentage for each formulation. For the amount of oil selected,
give sufficient flexibility to interfacial film such that it can change the minimum possible concentration of Smix should be used. Smix
its curvature to allow formation of nanoemulsion over a wide range ratios 1:0 and 1:4 were not used for making drug loaded formula-
of composition. In addition, co-surfactants also enhance the mobil- tions because of small NE area which implies decreased ability for
ity of hydrocarbon tails of oil thus allowing better penetration. On emulsification.
further raising the co-surfactant concentration i.e. Smix 1:4, the NE
area was again reduced most probably indicating formation of less
number of micelles thus it was not able to sufficiently reduce the
3.4. Thermodynamic stability of nanoemulsions (NE)
interfacial energy (Fig. 2E).
In order to prepare a stable NE, the effect of change in surfac-
NEs are thermodynamically stable systems and are formed
tant concentration (Tween 80) has been observed with respect to
at a particular concentration of oil, surfactant, and water, mak-
changes in NE area. When the Smix ratio was enhanced from 2:1 to
ing them stable and not subject to phase separation, creaming,
4:1, the NE area concomittantly increased (Fig. 2F–H). These results
or cracking. It is the thermostability that differentiates nano- or
suggest surfactants have the ability to reduce the interfacial energy
microemulsions from emulsions that have kinetic stability and
but only upto to a certain concentration (CMC). When the concen-
eventually phase-separate [18]. Thus the formulations were tested
tration exceeds the CMC, the capability is hindered without any
for their thermodynamic stability by checking their stability at low
effect on interfacial energy.
temperature and high shear. The formulations which passed the
thermodynamic protocol were namely 12:40:48 (1:1), 15:40:45
3.3. Selection of stable nanoemulsion (NE) (2:1), 18:40:42 (2:1), 15:35:50 (3:1) and 18:36:46 (4:1); these were
then coded as NE1, NE2, NE3, NE4 and NE5 respectively (Table 1).
From the pseudo-ternary phase diagram studies, placebo formu- These were also the ones that contained the minimum amount
lation was found sufficiently stable. Therefore our next step was to of surfactant which is a major criterion for selection of such for-
prepare drug loaded NE keeping the oil and Smix ratios same. The mulations. The rest of the formulations showed phase separation,
amount of oil should be sufficient enough to solubilise the dose turbidity, change in color or drug precipitation suggest instabil-
of curcumin in 1 ml of the NE. As per saturation solubility studies ity and were rejected. The composition of selected nanoemulsions
of curcumin in the oil phase (Labrafac PG: Triacetin, 1:1), 25 mg of (NE1-NE5) were given in Table 1.

Fig. 3. In-vitro skin permeation profile of curcumin from 5 different nanoemulsion formulations (NE1-NE5).
L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579 575

Table 2 Table 4
Permeability Parameters of Different nanoemulsions (NE1-NE5). Skin irritation studies of optimized nanoemulsion (NE2).

Formulation code Flux (␮g/cm2 /h) (n = 3) Permeability coefficient Rat Group Score after (days) Mean Score

NE1 75.306 ± 5.4 0.030 1 2 3 4 5 6 7


NE2 77.175 ± 7.2 0.031
NE3 107.1 ± 5.9 0.043 I 0 1 0 0 1 0 1 0.43
NE4 95.106 ± 9.2 0.038 II 1 0 0 0 0 1 0 0.29
NE5 92.733 ± 8.6 0.037 III 0 1 1 0 0 0 0 0.29

Table 5
Optimization of chitosan concentration for development of nanoemulsion gel.
3.5. Ex-vivo skin permeation study and skin deposition study
S.No. Percentage of chitosan Gel formation Spreadability
Ex-vivo skin permeation studies were performed to compare the 1. 1% Not formed Good
release of drug from five different NE formulations (NE1 –NE5 ), all 2. 2% Formed Good
having the same quantity of curcumin. Fig. 3 clearly demonstrates 3. 4% Formed Poor

the skin permeation of curcumin from various NEs. The flux values
abruptly increased from 75.3 ␮g/cm2 /h (NE1) to 107.1 ␮g/cm2 /h
in smaller in size as compared to the droplet size measured using
(NE3) followed by gradual decrease to 92.7 ␮g/cm2 /h (NE5), sug-
photon correlation spectroscopy.
gesting enhanced skin permeation shown by NE3 formulation
Viscosity measurements were carried out and average viscos-
(Table 2). Upon investigation, It was observed that NE2 and NE3
ity of NE2 was found to be 78.23 cps. It is noted that the viscosity
had same content of Tween 80 (2:1) but flux values found less in
of the formulations was low due to low amount of Tween 80 and
NE2 as compared to NE3, which may be due to difference in ratio
oil content, which is expected for nanoemulsion suggesting ease in
of oil and water composition used in formulation.
handling. Refractive index of the NE was determined using Abbes
Higher content of oil present in formulation NE3 was respon-
type refractometer. Thr mean value of refractive index for the for-
sible for higher flux values as lipophilic drugs are preferably
mulation was 1.404, suggesting isotropic nature of formulation. The
solubilized in o/w nanoemulsion. Since Tween 80 is a polysor-
pH values of the prepared formulations ranged from 6.2 to 6.3,
bate surfactant by nature, therefore it possesses the capability
which is considered acceptable as the skin pH is reported to be
to enhance drug permeation by penetrating into the intracellular
5.5-6.5 Therefore the formulation avoids the risk of irritation upon
regions of stratum corneum and eventually solubilise lipid compo-
application to the skin.
nents. However, this effect was observed only upto a certain extent.
As the amount of Tween 80 is increased further (NE4- 3:1, NE5- 4:1),
3.7. Skin irritation test
the rate of skin permeation fell gradually. This can be possibly due
to overall reduced Smix content, 35% (NE4) and 36% (NE5) vs 40%
Curcumin is a known irritant and in addition the formulation
in NE3. These findings have been supported by permeability coeffi-
contains varying amounts of surfactants making it necessary to
cient data for NE1 (0.03), NE2 (0.031), NE3 (0.043), NE4 (0.038) and
conduct skin irritation study. The test revealed a mean irritation
NE5 (0.037) which shows a similar pattern. Since the formulation is
score value of 0.34 for the optimized NE formulation (NE2). It has
designed for use in the treatment of wounds, more skin retention is
been previously reported that a value between 0 and 9 suggest
required rather than permeation. Therefore, skin deposition of the
non-irritant potential (Table 4) hence our formulation is safe and
curcumin in the five formulations (NE1-NE5) were investigated and
compatible for topical delivery to human skin [28].
were found in the range of 17.46–46.07%. Since highest skin depo-
sition was observed for formulation NE2 with 46.07% deposition
(Table 3), therefore it was selected for further studies. 3.8. Formulation and evaluation of nanoemulsion gel

On the basis of permeation and characterization studies, the


3.6. Characterization of nanoemulsion (NE) optimized formulation NE2 was selected to be formulated into gel.
Chitosan was selected for the preparation of gel due to its biocom-
Since our aim is to formulate NE therefore observing droplet size patible and non-toxic nature. It posesses an ability to form gels at
is of immense significance. By definition, a typical nanoemulsion acidic pH values, because it is hydrophilic and can retain water in
should have droplet sizes less than 100 nm and the results of our its structure. Gels containing 1%, 2% and 4% chitosan were prepared
study prove that nanoemulsions were formed. The average droplet and the best concentration was selected on the basis of visual obser-
size of nanoemulsion NE2 was found to be 84.032 nm with PDI vations. 2% gel was found to be suitable for gelling the NE because
0.232. Further TEM studies were conducted to determine the sur- of its desirable consistency, feel and ease of spreadability (Table 5).
face morphology of nanoemulsion. In the TEM positive image, the Additionally, 0.025% of cetyl pyridinium chloride was incorporated
nanoemulsions droplets appear circular. The light surroundings is into chitosan gel to incur anti-infectious properties.
due to opaque nature of oil phase whereas the darker area signifies The NEG was subjected to the study of various parameters for its
the embedded curcumin (Fig. 4). TEM image showed the presence of characterization (Table 6). The spreadability of the gel was found
spherical non-aggregated droplets within nanorange, which were to be 7.1 cm and viscosity was measured to be 504 ± 21.4 cps.

Table 3
Skin deposition studies of developed nanoemulsions (NE1-NE5).

Nanoemulsions Concentration (␮g/ml) Amount of drug in skin (␮g) (mean ± SD)a (n = 3) % of drug deposition

NE1 132.5 662.5 ± 22 26.5%


NE2 194.25 1811 ± 97 46.07%
NE3 112.8 564.0 ± 27 22.56%
NE4 87.3 436.5 ± 35 17.46%
NE5 94.45 472.25 ± 41 18.89%
576 L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579

Fig. 4. Transmission electron microscopic image of curcumin nanoemulsion.

Table 6
Comparative Evaluation.

Formulation Spreadability Drug content (mg) Droplet size (nm) Viscosity (cps) pH Cumulative Amount of drug
amount of drug retained in skin
permeated (␮g) (␮g)

NEG 7.1 ± 0.115 2.45 ± 0.008 504 ± 21.4 6.4 ± 0.121 1356.46 980.75 ± 88
NE2 – – 84.032 ± 0.023 78.23 ± 22.2 6.2 ± 0.224 1811.35 771.25 ± 67
Plain gel – – – – – 392.14 154.32 ± 15

(n = 3).

80 of chitosan which enmeshed the drug and precluded the release.


70 The thickening effect of chitosan gel has lent the formulation a bet-
% Cumulative drug permeated

ter spreadability. It also ensured that the drug deposition is higher,


60 nearing approximately 40%. This retention of curcumin in the skin
50 will ensure the hastening of re-epithelialisation process. Moreover
NE2
the presence of chitosan load with cetyl pyridinium chloride as
40
a bed within the midst of the wound would probably ensure an
30 NEG
infectious moist environment for fast healing of the wound. In addi-
20 control tion skin permeation of curcumin from NEG was compared with
plain curcumin gel. Enhanced skin permeation was observed on
10
using NEG (31.25%)/NE2 (46.07%) in contrast to plain gel (11.47%).
0 This significant difference can be attributed to small droplet size
0 5 10 15 20 25 30 of nanoemulsion which can permeate through skin barriers effi-
Time (hr) ciently. Numerous surfactant/co-surfactants have been evaluated
as penetration enhancer for the development of nanoemulsions
Fig. 5. Ex-vivo skin permeation through rat skin at different time intervals on appli-
cation of Contro, NE2 and NEG (n = 3). such as Tween 80 and PEG 400 which facilitate improved perme-
ation of curcumin in topically applied dosage forms. [18]. Since
curcumin is hydrophobic in nature therefore limited penetration is
These parameters signify sufficient viscosity that facilitates ease observed from plain gel. Therefore, it was found that NE have suc-
in application and spreading. Further the pH of the formulation cessfully enhanced skin permeation and deposition of curcumin by
was 6.4 ± 0.121, suggesting that the formulation is compatible to upto 6 fold.
human skin. The drug content was also evaluated since it is an
important factor which determines the amount of drug reaching 3.9. Physical stability studies
the skin. 98% (2.45 mg) drug content signifies good drug loading
capacity. The flux of nanoemulsion gel was 68.88 ␮g/cm2 /h, when Stability testing was carried out to investigate the effect of envi-
compared to that of nanoemulsion NE2 (76.05 ␮g/cm2 /h) is signifi- ronmental factors such as variations in temperature, humidity and
cantly lower suggesting limited skin permeation of curcumin from light on the formulation. A NEG should undergo physical and chem-
gel formulation (Fig. 5). Further the amount of curcumin retained in ical estimations to determine their stability. The gel formulation is
skin by gel formulation (980.75 ± 88 ␮g) is significantly higher than considered stable if there is no phase separation, turbidity or coa-
NE2 formulation (771.25 ± 67 ␮g). The reduction in permeation is lescence. For chemical stability, we have tested the drug content in
actually attributable to the gel formulation and thickening action the formulation at different time periods. The log% drug remaining
L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579 577

2.002 120
2
100
Log % drug remaining

1.998

% Wound Healing
1.996 80
1.994 30°C Control
60
1.992 40°C Placebo

1.99 50°C Treatment


40
1.988
20
1.986
0 20 40 60 80 100
0
Time (days)
Day3 Day6 Day9 Day12 Day15 Day18
Fig. 6. Stability studies of nanoemulsion gel (NEG), Log percentage remaining vs
Fig. 8. Incisional wound healing study in of nanoemulsion gel on albino rat model
time.
(n = 3).

1/T*1000
-3.56
dragged behind significantly. This suggests that treatment with
the cucumin NEG enhanced wound healing as compared with the
-3.58 3.05 3.1 3.15 3.2 3.25 3.3 3.35
groups not receiving it. It can be observed that placebo formulation
-3.6
was successful in healing wound in 15 days as compared to con-
-3.62 trol group (18 days). The wound healing properties of curcumin
-3.64 is mainly due its ability to enhance granulation tissue formation,
log K

-3.66 collagen deposition, tissue remodeling and wound contraction as


-3.68 reported in literature. Hence,this wound healing potential of NEG
was probably due to definite wound contraction [31]. It has been
-3.7
reported that chitosan acts a wound healing accelerator. The effi-
-3.72
cacy of chitosan for wound healing is mainly due to its acceleration
-3.74 of the infiltration of polymononuclear cells into the wound area,
stimulation of the migration of microphages, proliferation of fibrob-
Fig. 7. Arrhenius plot for the calculation of shelf-life.
lasts and the production of collagen [32]. For detailed mechanistic
insights on the wound healing activity of NEG, further studies are
for each temperature (30 ◦ C, 40 ◦ C and 50 ◦ C) was calculated and needed. A primary requirement for wound healing is to include a
plotted against time (Fig. 6). The degradation rate constant was moist and bacteria free environment. Availability of the antisep-
calculated by the following equation (Table 7): tic (cetyl pyridinium chloride) in the aqueous phase will prevent
chances of infection. On the contrary, enhanced wound healing
Slope = −K/2.303 effect observed with NEG formulation can be attributed to the com-
bined effect of chitosan and curcumin.
Arrhenius plot was drawn by plotting log K vs 1/T (Fig. 7) and
Additionally, the NE was able to facilitate permeation of cur-
value of K at 25 ◦ C (K25 ) was obtained by extrapolation. The shelf-
cumin to the underlying skin layers of the wound because of its
life (t0.9 ) i.e. time required for 10% degradation of the drug was
tendency to dissolve the superficial layers of skin lipids. They
calculated from the following equation: t0.9 = 0.1054/ K25. The shelf-
also provide better reservoir for a poorly soluble drug through
life of the formulation was found to be 1.58 years. The results shows
their capacity for enhanced solubilisation. An added advantage of
that the formulation is chemically and physically stable for 1.58
nanoemulsion, is the depot effect of curcumin in the skin which
years.
helps in residing the drug for longer periods of time [7]. These
3.10 In-vivo wound healing study
outcomes have been justified and explained in detail in skin per-
The suitability of any new formulation is best tested through its
meation studies. Since curcumin used alone can be highly irritant
in-vivo performance. Often the success of a formulation in in-vitro
to the animal because of high amounts of plyphenol, therefore it
conditions is not reproduced in the in-vivo situation and hence its
was not included in the study protocol [4].
testing in a suitable animal model before progressing for commer-
cialization is needed. The effect of the application of the formulation
on the healing of incisional wounds in wistar rats was studied. Per- 4. Conclusion
cent reduction in the wound area in control, placebo and treatment
groups is presented in Figs. 8 and 9. All wounds showed a reduction Our studies have clearly shown that curcumin nanoemulsion
in size over time, regardless of the group to which they belonged. was formulated which was proved by pseudo-ternary phase dia-
But the group treated with the NEG showed full wound closure by grams and TEM studies. Further the in-vitro studies proved the
the 12th day of the study whereas the control and placebo groups formulation was sufficiently thermodynamically and physically

Table 7
Degradation rate constant for curcumin in nanoemulsion gel at different temperatures.

Temperature ◦ C Slope*105 K*104 (days−1 ) Log K Absolute temperature 1/T 1/T *103

30 −8.73 2.010 −3.6968 0.003300 3.3003


40 −9.96 2.293 −3.6395 0.003189 3.189
50 −11.70 2.694 −3.5696 0.003091 3.0951
25 0.003350 3.3500
578 L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579

Fig. 9. Photographical representation of wound contraction rate at different time intervals for A- control, B- placebo, C- treatment groups.

stable. The optimised formulation containing 15% Labrafac PG: Tri- Acknowledgement
acetin (1:1) as the oil phase and 40% Tween 80 and PEG 400 as
surfactants and co-surfactants, respectively in a ratio of 2:1, were Authors are thankful to Department of Pharmaceutics, Faculty
subjected to ex-vivo and in-vivo studies. These studies have proved of Pharmacy, Jamia Hamdard University, New Delhi, India, for pro-
that nanoemulsion can be used for loading curcumin for the treat- viding essential facility in this research.
ment of wound healing.

4.1. Statistical analysis References

[1] A.J. Singer, R.A.F. Clark, Cutaneous wound healing, New Engl. J. Med. 341
All the results obtained were expressed as mean ± S.D using (1999) 738–746.
Graph Pad Software (Instat 3.06, USA) with more than three repli- [2] F. Roesken, E. Uhl, S.B. Curri, M.D. Menger, K. Messmer, Acceleration of wound
cates for each experiment. healing by topical drug delivery via liposomes, Langenbecks, Arch. Surg. 385
(2000) 42–49.
[3] S.D. Fitzmaurice, R.K. Sivamani, R.R. Isseroff, Antioxidant therapies for wound
Conflict of interest statement healing: a clinical guide to currently commercially available products, Skin
Pharmacol. Phys. 24 (2011) 113–126.
[4] D. Gopinath, M.R. Ahmed, K. Gomathi, K. Chitra, P.K. Sehgal, R. Jayakumar,
Authors report no conflict of interest of this work. Author is Dermal wound healing processes with curcumin incorporated collagen films,
responsible for content and the writing of the paper. Biomaterials 25 (2004) 1911–1917.
L. Thomas et al. / International Journal of Biological Macromolecules 101 (2017) 569–579 579

[5] G.S. Sidhu, A.K. Singh, D. Thaloor, K.K. Banaudha, G.K. Patnaik, R.C. Srimal, R.K. [18] F. Shakeel, S. Baboota, A. Ahuja, J. Ali, S. Shafiq, Celecoxib nanoemulsion for
Maheshwari, Enhancement of wound healing by curcumin in animals, Wound transdermal drug delivery: characterization and In vitro evaluation, J. Disp.
Repair Regen. 6 (1998) 167–178. Sci. Technol. 6 (2009) 834–842.
[6] C. Gong, Q. Wu, Y. Wang, D. Zhang, F. Luo, X. Zhao, Y. Wei, Z. Qian, A [19] D. Attwood, C. Mallon, G. Ktistis, C.J. Taylor, A study on factors influencing the
biodegradable hydrogel system containing curcumin encapsulated in micelles droplet size in nonionic oil-in-water microemulsions, Int. J. Pharm. 88 (1992)
for cutaneous wound healing, Biomaterials 34 (2013) 6377–6387. 417–422.
[7] A. Teichmann, S. Heuschkelb, U. Jacobia, G. Pressea, W. Sterrya, J. Lademanna, [20] M. Sakkinen, U. Seppala, P. Heinanen, M. Marvola, In vitro evaluation of
Comparison of stratum corneum penetration and localization of a lipophilic microcrystalline chitosan (MCCh) as gel-forming excipi ent in matrix
model drug applied in an o/w microemulsion and an amphiphilic cream, Eur. granules, Eur. J. Pharm. Biopharm. 54 (2002) 33–40.
J. Pharm. Biopharm. 67 (2007) 699–706. [21] K.A. Shah, A.A. Date, M.D. Joshi, V.B. Patravale, Solid lipid nanoparticles (SLN)
[8] S. Wang, F. Zheng, Y. Huang, Y. Fang, M. Shen, M. Zhu, X. Shi, Encapsulation of of tretinoin: potential in topical delivery, Int. J. Pharm. 345 (2007) 163–171.
amoxicillin within laponite-Doped poly(lactic-co-glycolic acid) nanofibers: [22] S.D. Mandawgade, V.B. Patravale, Development of SLNs from natural lipids:
preparation characterization, and antibacterial activity, ACS Appl. Mater. application to topical delivery of tretinoin, Int. J. Pharm. 363 (2008) 132–138.
Interfaces 4 (2012) 6393–6401. [23] F. Shakeel, S. Baboota, A. Ahuja, J. Ali, M. Aqil, S. Shafiq, Nanoemulsions as
[9] S. Wang, J. Zhao, F. Hu, X. Li, X. An, S. Zhou, Y. Chen, M. Huang, vehicles for transdermal delivery of aceclofenac, AAPS PharmSciTech 8 (2007)
Phase-changeable and bubble-releasing implants for highly efficient E1–E9.
HIFU-responsive tumor surgery and chemotherapy, J. Mater. Chem. B 4 (2016) [24] U.D. Kulkarni, R. Mahalingam, X. Li, I. Pather, B. Jasti, Effect of experimental
7368–7378. temperature on the permeation of model diffusants across porcine buccal
[10] M.K. Anwer, S. Jamil, E.O. Ibnouf, F. Shakeel, Enhanced antibacterial effects of mucosa, AAPS PharmSciTech 12 (2011) 579–586.
clove essential oil by nanoemulsion, J. Oleo Sci. 63 (2014) 347–354. [25] F. Shakeel, W. Ramadan, Transdermal delivery of anticancer drug caffeine
[11] E. Abd, S. Namjoshi, Y.H. Mohammed, M.S. Roberts, J.E. Grice, Synergistic skin from water-in-oil nanoemulsions, Colloids Surf. B Biointerfaces 75 (2010)
penetration enhancer and nanoemulsion formulations promote the human 356–362.
epidermal permeation of caffeine and naproxen, J. Pharm. Sci. 105 (2016) [26] D. Kumar, M. Aqil, M. Rizwan, Y. Sultana, M. Ali, Investigation of a
212–220. nanoemulsion as vehicle for transdermal delivery of amlodipine, Pharmazie
[12] H.P. Thakkar, A. Khunt, R.D. Dhande, A.A. Patel, Formulation and evaluation of 64 (2009) 80–85.
Itraconazole nanoemulsion for enhanced oral bioavailability, J. [27] E. Peira, E. Carlotti, C. Trotta, R. Cavalli, M. Trotta, Positively charged
Microencapsul. 32 (2015) 559–569. microemulsions for topical application, Int. J. Pharm. 346 (2008) 119–123.
[13] B. Baroli, M.A. López-Quintela, M.B. Delgado-Charro, A.M. Fadda, J. [28] N.J. Van Abbé, P.M. Dean, The measurement of antidandruff efficacy, Int. J.
Blanco-Méndez, Microemulsions for topical delivery of 8-methoxsalen, J. Dermatol. 14 (1975) 503–505.
Controll. Release 69 (2000) 209–218. [29] S. Ejaz, I. Chekarova, J.W. Cho, S.Y. Lee, S. Ashraf, S.W. Lim, Effect of aged garlic
[14] Y. Y.Yuan, J. Gao, L. Zhao, Mao Characterization and stability evaluation of extract on wound healing: a new frontier in wound management, Drug Chem.
␤-carotene nanoemulsions prepared by high pressure homogenization under Toxicol. 32 (2009) 191–203.
various emulsifying conditions, Food. Res. Int. 41 (2008) 61–68. [30] M. El-Badry, N. Haq, G. Fetih, F. Shakeel, Solubility and dissolution
[15] P. Puranjoti, R.T. Patil, P.D. Seth, G. Bommareddy, P. Dondeti, K. Egbaria, enhancement of tadalafil using self-nanoemulsifying drug delivery system, J.
Design and development of transdermal microemulsion for poorly water Oleo Sci. 63 (2014) 567–576.
soluble antifungal agents, J. Appl. Res. Clin. Exp.Ther. 2 (2002) 231–237. [31] D. Akbik, M. Ghadiri, W. Chrzanowski, R. Rohanizadeh, Curcumin as a wound
[16] J. Ali, N. Akhtar, Y. Sultana, S. Baboota, A. Ahuja, Antipsoriatic microemulsion healing agent, Life Sci. 116 (2014) 1–7.
gel formulations for topical drug delivery of babchi oil (Psoralea corylifolia), [32] K. Azuma, R. Izumi, T. Osaki, S. Ifuku, M. Morimoto, H. Saimoto, S. Minami, Y.
Methods Findings, Exp. Clin. Pharmacol. 30 (2008) 1–9. Okamoto Chitin, Chitosan: and its derivatives for wound healing: old and new
[17] S. Shafiq, F. Shakeel, S. Talegaonkar, F.J. Ahmad, R.K. Khar, M. Ali, Development materials, J. Funct. Biomater. 6 (2015) 104–142.
and bioavailability assessment of ramipril nanoemulsion formulation, Eur. J.
Pharm. Biopharm. 66 (2007) 227–243.

Anda mungkin juga menyukai