You are on page 1of 10

Original Research

Topical delivery of aceclofenac as


nanoemulsion comprising
excipients having optimum
1. Introduction
2. Materials and methods
emulsification capabilities:
3. Results and discussion preparation, characterization and
in vivo evaluation
4. Conclusion
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

Sandipan Dasgupta, Sanjay Dey, Supratik Choudhury &


Bhaskar Mazumder†

Dibrugarh University, Department of Pharmaceutical Sciences, Assam, India

Objective: The aim of the present study was to investigate the potential of a
nanoemulsion for topical delivery of aceclofenac using different excipients
having optimum emulsifying ability rather than their solubilizing capacity.
Methods: The oil-in-water nanoemulsions were prepared by screening the
excipients from the nanoemulsion region of pseudoternary phase diagram.
The prepared nanoemulsions were subjected to different thermodynamic
stability tests. The nanoemulsion formulations that passed thermodynamic
For personal use only.

stability tests were characterized for viscosity, droplet size, transmission


electron microscopy, refractive index and in vitro skin permeation. The
in vitro skin permeation profile of optimized nanoemulsion formulation
(NE31, containing 23.85% Polyoxy-35-castor oil, 7.95% PEG 400 and 13.6%
Triacetin) was compared with that of nanoemulsion gel (NG31) and marketed
gel formulation (HIFENAC GEL (HIG)). In vivo anti-inflammatory efficacy
studies were also carried out for NE31, NG31 and HIG.
Results: The significant (p < 0.001) increase in in vitro permeability and in vivo
anti-inflammatory efficacy of the NG31 formulation was observed as
compared with HIG formulation.
Conclusion: It can be concluded that the selection of surfactant and cosurfac-
tant on the basis of their emulsification capabilities other than the solubiliz-
ing capacity of drug is an important criterion for the formulation
of nanoemulsion.

Keywords: aceclofenac, emulsification capability, nanoemulsion, topical delivery

Expert Opin. Drug Deliv. (2013) 10(4):411-420

1. Introduction

In the last few years, the delivery of highly lipophilic drugs in the form of nanoe-
mulsion have gained a potential interest to most research scientists due to its certain
advantages like ease of formulation, non-toxicity and optimum permeable capacity
through skin.
Osteoarthritis (OA) is the most common disease after the age of 65 in about 60%
of men and 70% of women [1]. OA is primarily a non-inflammatory degenerative
joint disease characterized by progressive loss of articular cartilage, subchondrial
bone sclerosis, osteophyte formation, changes in the synovial membrane and an
increased volume of synovial fluid with reduced viscosity and hence, changed lubri-
cation properties. Current treatment options for OA are limited. They include

10.1517/17425247.2013.749234 © 2013 Informa UK, Ltd. ISSN 1742-5247, e-ISSN 1744-7593 411
All rights reserved: reproduction in whole or in part not permitted
S. Choudhury et al.

intra-articular (IA) injection of glucocorticoids and hyalur- The present research work was aimed to explore the feasibil-
onic acid (HA) preparations or symptomatic treatment with ity of nanoemulsion as a novel carrier system for topical
simple non-steroidal anti-inflammatory drugs (NSAIDs) [2]. application of aceclofenac for modulation of drug release.
Due to the localized nature of the disease, the IA injection The in vitro permeation of drug and anti-inflammatory
of glucocorticoids and HA seems to be an attractive approach activity of nanoemulsion gel were also evaluated.
for OA, but they provide only short-term pain relief and/
or often do not provide adequate pain relief and have no 2. Materials and methods
patient compliance [2].
On the other hand, rheumatoid arthritis (RA) is a wide- 2.1 Materials
spread chronic systemic inflammatory disease that is primarily Aceclofenac was obtained as gift sample from Cipla Ltd.
characterized by inflammation of the synovium with destruc- (Sikkim, India). Isopropyl myristate (IPM), oleic acid, castor
tion of cartilage and bone as the disease progresses. More oil, ethyl oleate, Tween 80 and Cremophore EL were pur-
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

severe disease may be associated with vasculitis, pericarditis, chased from HiMedia (Mumbai, India). Span 80 was pur-
pleural effusion, pulmonary interstitial fibrosis, peripheral chased from Loba Chemicals (Mumbai, India). Glycerol
neuropathies, subcutaneous and pulmonary nodules and triacetate (Triacetin) was purchased from E-Merck (Mumbai,
scleritis [3,4]. Since there is no cure, symptomatic treatment India). HIFENAC GEL (HIG) (30 g; batch no. M12 02/12;
is the only choice to reduce the pain and inflammation. manufactured by Intas Pharmaceutical Ltd., Ahmedabad,
Formulations of NSAIDs are the first-choice remedies which India) was purchased from local pharmacy. All other chemicals
would fulfill the need of providing significantly long used in the study were of analytical grade.
remission of pain with improved patient compliance.
Aceclofenac is an NSAID, which is used for the treatment 2.2 Screening of oil
of OA and RA [5-7]. It comes under the category II drug The solubility of aceclofenac in various oils was determined
under Biopharmaceutical Classification System (BCS). It by adding an excess amount of drug in 5 ml of selected oils
has an intermediate half-life of 3 -- 4 h and it requires fre- (castor oil, Triacetin, IPM, ethyl oleate, oleic acid) and kept
For personal use only.

quent oral dosing because of its short half-life. It undergoes under magnetic stirring at temperature of 25 ± 1.0 C for
substantial first-pass metabolism. Oral administration of ace- 72 h. The equilibrated sample was centrifuged at 3000 rpm
clofenac may lead to various side effects like gastrointestinal for 15 min to separate the undissolved drug. The supernatant
ulcer and bleeding on chronic use which results in anemic was taken and filtered through 0.45 µm membrane filter. The
condition. Therefore, the topical delivery of aceclofenac concentration of aceclofenac in oil was determined using
would be possible alternative offering distinct advantages UV-Visible spectrophotometer (UV-1700, Shimadzu, Tokyo,
such as elimination of the absorption variable rate, first- Japan) at 273 nm.
pass intestinal and hepatic metabolism inherent with oral
dosing and delivering the drug directly to the inflamed 2.3 Screening of surfactants
site and thereby, producing high local concentrations and The emulsification ability of the surfactants was evaluated by
avoiding the side effects [8]. adding 300 mg of surfactant into 300 mg of the selected oily
The designing of a topical drug delivery system of aceclofe- phase. The mixture was gently homogenized at 45 -- 60 C.
nac could not only increase payload of drug locally and The isotropic mixture of 50 mg was accurately weighed and
improve the release of drug for a prolonged period of time diluted with distilled water to yield a fine emulsion volume
but also reduce the risk of systemic toxicity. Nanoemulsions, of 50 ml. The ease of formation of emulsion was monitored
owing to their small size, provide a large interfacial area for by noting the number of volumetric flask inversions required
rapid drug release, thereby enhancing bioavailability, reducing to give uniform emulsion. The emulsions were allowed to
dose size, providing consistent temporal profiles of drug stand for 2 h to note for any change in turbidity through
absorption and protecting the drugs from the hostile environ- visual observation and their transmittance was assessed at
ment of the body. In addition, nanoemulsions could be used 273 nm by colorimeter (6051 Jenway, UK) using distilled
as a reservoir for sustained release of drug for prolonged water as blank.
period of time, thus avoiding high concentration of drug in
the blood [9,10]. Though, nanoemulsion formulations of ace- 2.4 Screening of cosurfactant
clofenac have been already reported and the selection of ingre- The cosurfactant was selected by mixing 100 mg of cosurfac-
dients for the formulation of nanoemulsion was made based tant in 200 mg of the previously selected surfactant and the
on their solubilizing capacity rather than their emulsification surfactant--cosurfactant mixture (Smix) was added to the
ability [11-13]. The present research work has enlightened the selected oily phase. The mixture was gently heated at
selection of ingredients for the formulation of nanoemulsion 45 -- 60 C for homogenizing the components. The 50 mg
based on their emulsification capacity rather than their of isotropic mixture was accurately weighed and diluted
solubilizing properties and their potential on aceclofenac with distilled water to 50 ml to yield fine emulsion. The
permeability through topical route. ease of formation of emulsions was monitored by noting the

412 Expert Opin. Drug Deliv. (2013) 10(4)


Topical delivery of aceclofenac as nanoemulsion comprising excipients having optimum emulsification capabilities

number of volumetric flask inversions required to give taken for heating and cooling cycle. Six cycles between
uniform emulsion. The resulting emulsions were observed refrigerator temperature of 4 C and 45 C with storage at
visually for the relative turbidity. The emulsions were allowed each temperature of not less than 48 h were studied. Those
to stand for 2 h to note for any change in turbidity and their formulations which were stable at these temperatures were
transmittance was assessed at 273 nm by colorimeter using subjected to freeze-thaw cycle test.
distilled water as blank. As the ratio of cosurfactants to surfac- iii) Freeze-thaw cycle: Three freeze-thaw cycles were done
tant/s is the same, the turbidity of resulting nanoemulsions between -21 C and +25 C with storage at each tempera-
will help in assessing the relative efficacy of the cosurfactants ture for not less than 48 h for the formulations.
to improve the nanoemulsification ability of surfactant/s.
Those formulations which passed these thermodynamic
2.5 Pseudoternary phase diagram stability tests were selected for further studies.
On the basis of the solubility studies of drug, Triacetin,
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

Polyoxy-35-castor oil and PEG 400 were selected as the oil 2.8 Characterization of nanoemulsion
phase, surfactant and cosurfactant, respectively. Distilled 2.8.1 Droplet size analysis
water was used as an aqueous phase. Surfactant and cosurfac- The globule size of the nanoemulsion was determined by
tant (Smix) were mixed in different weight ratios (1:3, 1:2, 1:1, photon correlation spectroscopy. The nanoemulsions (0.1 ml)
2:1 and 3:1). These Smix ratios were chosen in increasing con- were diluted with 50 ml of water in a volumetric flask and gently
centration of surfactant with respect to cosurfactant and mixed by inverting the flask. Measurement was done using a
increasing concentration of cosurfactant with respect to sur- Zetasizer 1000 HS (Malvern Instruments, UK). Light scattering
factant for detailed study of the phase diagrams. For each was monitored at 25 C at a 90 angle. Droplet size and
phase diagram, oil and Smix at specific ratio was mixed thor- polydispersity index were obtained as the average of
oughly in different weight ratios from 1:9 to 9:1 in different three measurements.
glass vials. Sixteen different combinations of oil and Smix,
1:9, 1:8, 1:7, 1:6, 1:5, 2:8 (1:4), 1:3.5, 1:3, 3:7 (1:2.3), 1:2,
2.8.2Viscosity
For personal use only.

4:6 (1:1.5), 5:5 (1:1), 6:4 (1:0.7), 7:3 (1:0.43), 8:2 (1:0.25),
The viscosity of the formulations (0.5 ml) was determined
9:1 (1:0.1), were made so that maximum ratios were covered
without dilution using Brookfield DVE viscometer (Brook-
for the study to delineate the boundaries of phases precisely
field Engineering Laboratories, Inc., Middleboro, MA,
formed in the phase diagrams. Pseudoternary phase diagrams
USA) using spindle no. 63 at 25 ± 0.5 C. The software
were developed using aqueous titration method. Slow titra-
used to calculate the viscosity was Rheocalc V2.6. Average
tion with aqueous phase was performed for each weight ratio
and standard deviation of three data at shear rate of 120.0 s-1
of oil and Smix and visual observations were made for trans-
were reported.
parent and easily flowable oil-in-water (o/w) nanoemulsions.
The physical state of the nanoemulsion was marked on a
pseudo-three-component phase diagram with one axis repre- 2.8.3 Transmission electron microscopy
senting aqueous phase, the other representing oil and the third The surface topography of the nanoemulsions were studied
representing a mixture of surfactant and cosurfactant at fixed using transmission electron microscopy (TEM). A JEMCX
weight ratios. 100II operating at 200 kV capable of point-to-point resolu-
tion was used. A combination of bright-field imaging at
2.6 Selection of nanoemulsion formulations increasing magnification and of diffraction modes was used
From each phase diagram constructed, different formulas to reveal the form and size of the nanoemulsion. To perform
were selected from the nanoemulsion region so that the drug the TEM observations, the nanoemulsion formulations were
could be incorporated into the oil phase; 1.5% (m/m) of diluted hundred times with water. A drop of the diluted nano-
aceclofenac, which was kept constant in all the selected emulsion was directly deposited on the holey film grid and
formulations, was dissolved in the oil phase of nanoemulsion observed after drying.
formulation. Selected formulations were subjected to different
dispersion stability tests. 2.8.4Refractive index
The refractive index of placebo formulations and drug-loaded
2.7Thermodynamic stability studies formulations was determined using an Abbes-type refractometer
To overcome the problem of metastable formulation, thermo- (Macro Scientific Works, Delhi, India).
dynamic stability tests were performed, which were as follows:
2.8.5In vitro skin permeation studies
i) Centrifugation: Selected formulations were centrifuged In vitro permeation studies were carried out on modified
at 3500 rpm for 30 min. Keshary-Chien diffusion cell with an effective diffusional
ii) Heating and cooling cycle: Those formulations that did area of 3.14 cm2 and 100 ml receiving chamber capacity,
not show any phase separation on centrifugation were using rat abdominal skin. The full thickness of rat skin was

Expert Opin. Drug Deliv. (2013) 10(4) 413


S. Choudhury et al.

Table 1. Solubility of aceclofenac in different oils*. (Jss, µg/cm2/h) [14] of aceclofenac was calculated from the
slope of the plot using linear regression analysis. The perme-
Oil Solubility (mg/ml) ability co-efficient (Kp) of the drug through the membrane
was calculated using the following equation [15]:
Castor oil 45.13 ± 0.97
Ethyl oleate 22.19 ± 0.16 Jss
IPM 3.42 ± 0.10 Kp =
Oleic acid 4.70 ± 0.05 C
Triacetin 49.89 ± 0.18
Where, C is the initial concentration of the drug in the
*Mean ± SD, n = 3. donor compartment.
IPM: Isopropyl myristate.
The penetration-enhancing effect was calculated in terms of
enhancement ratio (Er) by using the following equation:
excised from the abdominal region and hair were removed with
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

an electric clipper. The subcutaneous tissue was removed surgi- Jss of nanoemulsion formulation
Er =
cally and the dermis side was wiped with isopropyl alcohol to Jss of control (HIG)
remove adhering fat. The cleaned skin was washed with dis-
tilled water and stored at -21 C until further use. The skin 2.9 Skin irritation study
was brought to room temperature and mounted between the Skin irritation study was performed on four Wistar albinos.
donor and receiver compartment of Keshary-Chien diffusion About 5 cm2 area of hair from the back side were depleted
cell where the stratum corneum side was facing the donor com- with the help of depilatories and area was marked on both the
partment and the dermal side was facing the receiver compart- side. One side served as control while other as test. After 24 h,
ment. The receiver chamber was filled with methanolic nanoemulsion dispersion (NE31) and nanoemulsion gel for-
phosphate buffer saline pH 7.4 (30:70%, v/v). The receiver mulation (NG31) were applied (500 mg/rat). Once a day for
fluid was stirred with magnetic stirrer at a speed of 100 rpm 7 days observation were made for any sensitivity and the
and the temperature was maintained at 37 ± 1 C. One milliliter reaction if any was graded as: A: no reaction; B: slight, patchy
For personal use only.

of nanoemulsion formulation (1.5% mg/ml aceclofenac) or 1 g erythema; C: moderate but patchy erythema; D: moderate
of aceclofenac gel (1.5% mg/g) was placed into the donor com- erythema; E: severe erythema with or without edema.
partment and sealed with paraffin film to provide occlusive
conditions. Samples were withdrawn at regular intervals (0.5, 2.10 In vivo efficacy study
1, 2, 4, 8, 12, 18, 24 h), filtered through 0.45 µm membrane The anti-inflammatory activity was evaluated using the
filter and analyzed for drug content by UV-Visible carrageenan-induced hind paw edema method developed by
spectrophotometer (UV-1700) at 273 nm. Winter [18]. Wistar albino rats (six rats each group) of either
sex weighing 120 -- 150 g were used for these studies. Prior
2.8.6 Preparation of nanoemulsion gel formulation to the experiment, the approval was taken from the Institu-
On the basis of permeation study, nanoemulsion formulation tional Animal Ethical Committee, M.K.C.G. Medical College
of NE31 was formulated to gel formulation as it shows highest and Hospital, Odisha, India. All the rats were divided into four
permeability through rat skin. One gram of Carbopol 934P groups: Group I: served as control; Group II: rats were treated
was dispersed in sufficient quantity of distilled water. After with HIG; Group III: rats were treated with nanoemulsion
complete dispersion, the solution was kept in dark for 24 h (NE31) and Group IV: rats were treated with nanoemulsion
for complete swelling of Carbopol 934P. The aceclofenac- gel (NG31). The animals were housed in institutional animal
loaded nanoemulsion (NF31) was slowly added to the viscous house under standard conditions with free access to food and
solution of Carbopol 934P under magnetic stirring. The 1% water. Inflammation was induced by sub-plantar carrageenan
w/v of Carbopol 934P was used for the formulation of gel. injection and after 1 h, formulations were applied topically
The pH was adjusted between 6 and 8 using 0.5 ml of trietha- on the inflamed paw of rats by gently rubbing with index fin-
nolamine and 1 ml of PEG 400 was incorporated to prevent ger and the volume of the paw was measured. The thickness of
the evaporation of moisture. The nanoemulsion gel formula- the paw was measured using a digital plethysmometer (Orchid
tion was kept at ambient condition after which in vitro perme- Scientifics, Nasik, India) before injection (0 h) and at regular
ation study was performed using similar procedure as intervals (1, 2, 4, 8, 12, 24 h) after injection. The amount of
mentioned in Section 2.8.5. The in vitro permeation study paw swelling was determined for 24 h and expressed as percent
was also carried out for marketed gel formulation (HIG). edema relative to the initial hind paw volume. Percent inhibi-
tion of edema produced by each formulation-treated group
was calculated against the respective control group.
Permeation data analysis
2.8.7
The permeation profiles were constructed by plotting the 2.11 Statistical analysis
cumulative amount of aceclofenac permeated per unit dialysis Statistical analysis of anti-inflammatory activity was done by
membrane area (µg/cm2) versus time. The steady-state flux means of one-way ANOVA (analysis of variance) followed

414 Expert Opin. Drug Deliv. (2013) 10(4)


Topical delivery of aceclofenac as nanoemulsion comprising excipients having optimum emulsification capabilities

Table 2. Emulsification capability of surfactants and cosurfactants.

Surfactants Maximum number % Cosurfactants + Maximum number %


of inversions* Transmittance* Cremophor EL of inversions* Transmittance*

Cremophor EL 21 ± 2 90 ± 4 PEG 400 11 ± 1 98 ± 2


Span 80 42 ± 3 15 ± 2 Span 80 36 ± 4 42 ± 3
Tween 80 37 ± 2 47 ± 2 Tween 80 29 ± 2 64 ± 2
PEG 400 30 ± 1 62 ± 3

*Mean ± SD, n = 3.
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

S:CoS = 1:1 S:CoS = 1:2 S:CoS = 1:3


Smix Smix
A. B. Smix C.
10 10
90 10 90
90
20 20
80 20 80
80
30 30 30
70 70 70
40 40 40
60 60 60
50 50 50
50 50 50
60 60 60
40 40 40
70 70 70
30 30 30
80 80 80
20 20 20
90 90 90
10 10 10
Aqueous Oil Aqueous Oil Aqueous Oil
10 20 30 40 50 60 70 80 90 10 20 30 40 50 60 70 80 90 10 20 30 40 50 60 70 80 90
For personal use only.

S:CoS = 2:1 S:CoS = 3:1


Smix Smix
D. E. 10
10 90
90
20
20 80
80
30
30 70
70
40
40 60
60
50 50
50
50
60 60
40
40
70 70
30 30
80 80
20 20
90 90
10 10

Aqueous Oil Aqueous Oil


10 20 30 40 50 60 70 80 90 10 20 30 40 50 60 70 80 90

Figure 1. Pseudoternary phase diagrams showing the o/w nanoemulsion (shaded area) regions of Triacetin (oil), Cremophore
EL (surfactant), PEG 400 (cosurfactant) at different Smix ratios A) Smix 1:1, B) Smix 2:1, C) Smix 3:1, D) Smix 2:1 and E) Smix 3:1.

by Dunnett’s t-test; p value < 0.05 was considered oil [16,19]. The solubility of aceclofenac in different oils is
statistically significant. presented in Table 1. Among the various oils, the solubility of
aceclofenac was found to be highest in Triacetin (49.894 ±
3. Results and discussion 0.184 mg/ml). Therefore, Triacetin was selected as the oil phase
for the development of nanoemulsion formulation.
3.1 Selection of excipients The selection of surfactant or cosurfactant in the further
The important criteria for the selection of excipients for the study was governed by their ability to reduce interfacial ten-
formulation of nanoemulsion should be pharmaceutically sion to a small value to aid the dispersion process during the
acceptable, non-sensitizing and non-irritating to the skin preparation of the nanoemulsion. The emulsification capabil-
and should fall into the category of generally regard as safe ity of different surfactant and cosurfactant is presented
(GRAS) [17]. The selection of suitable oil having maximum in Table 2. These studies indicated that Polyoxy-35-castor
solubilizing capacity is very important, as the drug in the nano- oil had very good ability to emulsify aceclofenac followed by
emulsion present in a solubilized form and the drug loading in PEG 400, Tween 80 and Span 80. The hydrophile--lipophile
the nanoemulsion is greatly influenced by the solubility in balance (HLB) values of the surfactants used in the

Expert Opin. Drug Deliv. (2013) 10(4) 415


S. Choudhury et al.

Table 3. Composition of selected nanoemulsion formulations.

Formulation Oil:Smix ratio Smix ratio Components (% m/m)

Oil Surfactant Cosurfactant Water

NE11 1:3.03 1:1 7.7 11.7 11.7 68.9


NE12 1:3.5 1:2 7.4 8.6 17.3 66.7
NE13 1:2.34 1:3 6.6 3.9 11.6 77.9
NE21 1:4.95 2:1 11.11 36.7 18.3 33.89
NE31 1:2.33 3:1 13.6 23.9 7.9 54.6

Smix ratios were chosen to reflect increasing concentrations of


Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

cosurfactant with respect to surfactant and increasing concen-


trations of surfactant with respect to cosurfactant for detailed
study of the phase diagrams in the nanoemulsion formation.
The shaded portion of the phase diagram indicated the
nanoemulsion region and it was increased with an increase
in the concentration of surfactant (Figure 1). The maximum
and minimum nanoemulsion region was obtained with the
Smix ratio of 3:1 and 1:1, respectively. As the surfactant
concentration was increased in the Smix ratio from 2:1 to
3:1, a higher nanoemulsion region was observed, perhaps
because of further reduction of the interfacial tension, increas-
ing the fluidity of the interface, thereby increasing the entropy
For personal use only.

of the system. There may be greater penetration of the oil


phase in the hydrophobic region of the surfactant mono-
Figure 2. Transmission electron microscopic image of
mers [20,21]. The nanoemulsion region formed by Smix ratio
aceclofenac nanoemulsion.
of 4:1 was very small which may be due to absence of
sufficient quantity of cosurfactant. Hence, the Smix ratio of
investigation were above 10 except for Span 80. An important
4:1 was avoided.
criterion for selection of the surfactants is that the HLB value
of the surfactant should be greater than 10 to form
o/w nanoemulsion. In the present study, the authors have 3.3 Selection of nanoemulsion formulations
selected Polyoxy-35-castor oil as surfactant. The addition of Aceclofenac is a highly lipophilic drug, and its physicochemi-
cosurfactant is necessary to achieve transient negative interfa- cal properties suggest that it has good potential for transdermal
cial tension and fluid interfacial film. The cosurfactant drug delivery. Therefore, the composition of nanoemulsion
decreases the bending stress of interface and allows the interfa- formulation should be such that it must be free from skin irri-
cial film sufficient flexibility to take up different curvatures tation. It is well known that large amounts of surfactants cause
required to form nanoemulsion over a wide range of composi- skin irritation [22,23]; therefore, it is very important to deter-
tion. Among the different cosurfactants tested to evaluate the mine the surfactant concentration properly and use the
emulsification capacity with Cremophore EL, the PEG optimum concentration of surfactant in the formulation.
400 was found to have more emulsification capacity than The pseudoternary phase diagrams were carefully observed
Span 80 and Tween 80 (Table 2). Therefore, PEG 400 was and from them the formulations in which the amount of oil
selected as cosurfactant for the preparation of nanoemulsion. phase completely solubilized the drug and which could accom-
modate the optimum quantity of Smix and distilled water
3.2 Construction of pseudoternary phase diagram were selected for the study. The composition of selected
The construction of pseudoternary phase diagram makes it formulations is shown in Table 3.
easy to determine the range of concentrations of components
at which nanoemulsion form. Phase diagrams were con- 3.4 Thermodynamic stability tests
structed to determine the individual component ratio for the Nanoemulsions are thermodynamically stable systems and are
formulation of an o/w emulsion consisting of Triacetin, formed at a particular concentration of oil, surfactant and
Cremophore EL and PEG 400 as oil phase, surfactant and water, which make them stable to prevent phase separation,
cosurfactant, respectively. creaming or cracking (Table 4). Therefore, the different
Polyoxy-35-castor oil and PEG 400 were mixed in different formulations were subjected to stability studies by using
volume ratios (3:1, 2:1, 1:1, 1:2, 1:3) to form the Smix. These centrifugation, heating-cooling cycle and freeze-thaw cycle.

416 Expert Opin. Drug Deliv. (2013) 10(4)


Topical delivery of aceclofenac as nanoemulsion comprising excipients having optimum emulsification capabilities

Table 4. Thermodynamic stability studies (centrifugation, heating-cooling cycle and freeze-thaw cycle), droplet
size, polydispersity index, viscosity, refractive index of fresh and placebo different nanoemulsion formulations.

Code Centrifugation Heating-cooling Freeze-thaw Droplet PDI‡ Viscosity Refractive index*


cycle cycle size (nm)* (mPa s)*
Fresh Placebo

NE11 ü ü ü 58.17 0.228 130.61 ± 0.31 1.359 ± 0.002 1.353 ± 0.002


NE12 ü ü ü 64.75 0.431 112.80 ± 0.21 1.372 ± 0.009 1.374 ± 0.004
NE13 ü ü - Not performed
NE21 ü ü ü 41.45 0.224 221.42 ± 0.22 1.389 ± 0.010 1.384 ± 0.001
NE31 ü ü ü 39.48 0.230 339.51 ± 0.31 1.375 ± 0.003 1.372 ± 0.002

*Mean ± SD, n = 3.
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

z
Polydispersity index.
ü : Test passed; -: Test failed.

causes the interfacial film to condense and stabilize, while


Table 5. Permeability parameters of different the cosurfactant causes the film to expand. The polydispersity
nanoemulsion formulations. values of all the formulations were found to be within the
range of 0.22 -- 0.43. The polydispersity value less than
Formulation Jss (mg/cm2/h)‡ Kp  10-2 (cm/h)‡ Er
0.30 indicated the uniformity of droplet size.
HIG* 43.67 ± 2.11 0.29 ± 0.01 -
NE31 254.90 ± 1.25 1.70 ± 0.01 5.84 3.5.3 Viscosity determination
NE21 204.03 ± 4.65 1.36 ± 0.03 4.67
The viscosity of formulation NE31 was found to be the high-
For personal use only.

NE11 187.27 ± 3.26 1.25 ± 0.02 4.29


NE12 162.03 ± 3.67 1.08 ± 0.02 3.71 est (339.50 ± 0.31 mPa s) compared with other formulations
NG31 199.60 ± 6.93 1.33 ± 0.05 4.57 (Table 4). This may be due to decreased concentration of
cosurfactant. This deferred from one characteristic of nanoe-
*HIG: HIFENAC GEL was used as control. mulsion which is that they are usually of low viscosity.
z
Mean ± SD, n = 3.
Er: Enhancement ratio; Jss: Steady-state flux; Kp: Permeability coefficient.
Instead, a hypothesis can be put forward that due to the
higher viscosity of the nanoemulsions the droplets were not
able to coalesce and thus retained their nanosize. The viscosity
of the formulation NE12 was lower as compared with formu-
Only those formulations that survived thermodynamic lation NE11. This may be due to the lower oil concentration.
stability tests were selected for further study.
3.5.4Refractive index
3.5 Characterization of nanoemulsions The values of the refractive index of drug-loaded formulations
3.5.1 Transmission electron microscopy and placebo formulations are given in Table 4. It was found
The nanoemulsion appeared dark and with bright surroundings that there were no significant differences of refractive index
(Figure 2). The droplet size ranged between 29 and 71 nm and when drug-loaded formulations were compared with placebo
was in agreement with the droplet size distribution measured formulations. Therefore, it can be concluded that the nanoe-
using photon correlation spectroscopy (Table 4). mulsion formulations were not only thermodynamically sta-
ble but also chemically stable and remained isotropic; thus,
3.5.2 Droplet size analysis there were no interactions between nanoemulsion excipients
The droplet size decreased with the increase in oil concentra- and drug.
tion in the formulation (Table 4). The droplet size of formu-
lation NE31, containing 13.6% of oil was 39.48 ± 5.06 nm, 3.5.5 In vitro skin permeation studies
which was lower as compared with other formulations. There The permeation ability of the various nanoemulsion formula-
was only a slight difference in the mean droplet size of formu- tions (NE11, NE12, NE21 and NE31), nanoemulsion gel
lations NE31 and NE21, which may be due to the increase in (NG31) and aceclofenac gel (HIG) were evaluated using
the concentration of Smix in NE31. The droplet size in the in vitro permeation experiments. The permeation parameters
formulation of NE12 was more as compared with formula- of nanoemulsion formulations, nanoemulsion gel and
tion NE11, which may be due to the increased concentration aceclofenac gel are presented in Table 5. The permeation profiles
of cosurfactant. This result is in accordance with the report of NE11, NE12, NE21, NE31, NG31 and aceclofenac gel
that the addition of surfactant to nanoemulsion systems through rat skins are shown in Figure 3. In vitro skin permeation

Expert Opin. Drug Deliv. (2013) 10(4) 417


S. Choudhury et al.

30

Amount of aceclofenac permeated (mg cm-2)


25

20

15

10
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

0
0 2 4 6 8 10 12 14 16 18 20 22 24
Time (h)

NE31 NE21 NE11 HIG NG31

Figure 3. In vitro skin permeation profile of aceclofenac (mean ± SD) from nanoemulsion (NE11, NE21 and NE31),
nanoemulsion gel (NG31) and aceclofenac gel (HIFENAC GEL, HIG) (mean ± SD; n = 3).
For personal use only.

Table 6. Anti-inflammatory effects of nanoemulsion (NE31), nanoemulsion gel (NG31) and aceclofenac gel (HIG).

Time (h) In vivo performance Group

I II III IV

1 Edema* (%) 36.50 ± 0.02 34.09 ± 0.02z 29.90 ± 0.01z 33.16 ± 0.02z
Inhibition (%) - 6.61 18.09 9.15
2 Edema* (%) 44.47 ± 0.02 38.89 ± 0.02z 28.39 ± 0.02z 30.89 ± 0.01z
Inhibition (%) - 12.56 36.16 30.55
4 Edema* (%) 46.79 ± 0.02 36.62 ± 0.03z 23.12 ± 0.02z 27.59 ± 0.02z
Inhibition (%) - 21.74 50.59 41.02
8 Edema* (%) 47.81 ± 0.01 36.87 ± 0.02z 18.09 ± 0.02z 22.28 ± 0.02z
Inhibition (%) - 22.89 62.17 53.41
12 Edema* (%) 43.70 ± 0.02 28.28 ± 0.02z 14.82 ± 0.02z 18.73 ± 0.02z
Inhibition (%) - 35.28 66.08 57.13
24 Edema* (%) 37.28 ± 0.02 21.72 ± 0.02z 10.05 ± 0.02z 12.91 ± 0.03z
Inhibition (%) - 41.74 73.04 65.36

Group I: Control group; Group II: Animals treated with aceclofenac gel (HIG); Group III: Animals treated with nanoemulsion (NE31); Group IV: Animals treated
with nanoemulsion gel (NG31).
*Mean ± SD, n = 6.
z
Edema rate (%) values were statistically significant from the saline control using one-way ANOVA followed by Dunnett’s t-test at p < 0.001.
ANOVA: Analysis of variance; HIG: HIFENAC GEL.

rate had the highest rate in the formulation NE31 as compared permeation profiles. The skin permeation profile of
with the aceclofenac gel. The flux (Jss) and permeability coeffi- NE31 was significantly (p < 0.05) different from the skin per-
cient (Kp) of NE31 and NG31 were found to be 254.90 ± meation profile of NE21 but the skin permeation profiles of
1.25 µg/cm/h, 1.70  10-2 ± 0.01 cm/h and 199.60 ± NE21 and NG31 were not significantly different. The signif-
6.93 µg/cm/h, 1.33  10-2 ± 0.05 cm/h. The enhancement of icant (p < 0.05) difference in the aceclofenac permeation
the flux of aceclofenac was increased 5.84 and 4.57 times between nanoemulsion formulation and aceclofenac gel might
for NE31 and NG31, respectively as compared with commer- be due to the smallest size of internal phase droplets [24]. The
cial gel formulation of aceclofenac. Formulations NE11, required flux for aceclofenac was approximately 197.54 µg/
NE12, NE21 and NG31 showed an intermediate skin cm/h and was obtained by formulation NG31 (199.60 ±

418 Expert Opin. Drug Deliv. (2013) 10(4)


Topical delivery of aceclofenac as nanoemulsion comprising excipients having optimum emulsification capabilities

6.93 µg/cm/h). In order to reach the required flux, the area NE31. The enhanced anti-inflammatory effects of formula-
has to be decreased up to 3.11 cm2. The results of aceclofenac tion NE31 could be due to the enhanced permeation of
permeation from NG31 through the rat abdominal skin con- aceclofenac through the skin.
firmed that aceclofenac was permeated through the rat skin
and hence could possibly permeate through the human skin. 4. Conclusion

3.6 Skin irritation study On the basis of lowest droplet size, lowest polydispersity,
The skin irritation study of nanoemulsion dispersion (NE31) optimum viscosity, optimum surfactant and cosurfactant
and gel formulation (NG31) were performed to evaluate the concentration highest skin permeation and highest in vivo
safety of the optimized nanoemulsion formulation. The anti-inflammatory potential, the authors have selected
mean values of skin irritancy score following 7 days application NE31 as optimized formulation, which contained Triacetin
for formulation NE31 and NG31 were found to be 1.89 ± (13.6%), Cremophore EL (23.9%), PEG 400 (7.9%) and dis-
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

0.45 and 1.9 ± 0.5, respectively. Van-Abbe et al. reported tilled water (54.6%). The surfactant (Cremophore EL) and
that a value between 0 and 9 indicates that the applied formu- cosurfactant (PEG 400) were selected on the basis of their
lation is generally not an irritant to human skin [25]. This value highest emulsification capacity. Therefore, it can be concluded
concluded that the optimized nanoemulsion formulations that the selection of surfactant and cosurfactant on the basis of
were safe for topical delivery of aceclofenac. their emulsification capabilities other than solubilizing capac-
ity of drug is an important criterion for the formulation of
3.7 In vivo efficacy study nanoemulsion. From the in vitro and in vivo data it can also
Based on optimum skin permeation and lowest droplet size be concluded that nanoemulsion formulation NE31 has great
formulation, NE31 was chosen for the study of in vivo anti- potential for the transdermal delivery of aceclofenac.
inflammatory effects. The percentage inhibition after 12 h
administration was found to be high for NE31, that is, Declaration of interest
73.04% as compared with HIG (41.74%); this difference
For personal use only.

was significant (p < 0.01). The percent inhibition value for We are thankful to University Grants Commission (UGC),
formulations NG31 was found to be 65.36% (Table 6); the file no. 37-44/2009(SR)(ASM) for their financial support
difference is significant when compared with formulation for this work.

Bibliography
1. Sarzi-Puttini P, Cimmino MA, Scarpa R, E2 production following its intracellular 11. Delgado-Charro MB, Iglesias-Vilas G,
et al. Osteoarthritis: an overview of the conversion into cyclooxygenase Blanco-Mendez J, et al. Delivery of a
disease and its treatment strategies. inhibitors. Eur J Pharmacol hydrophilic solute through the skin from
Semin Arthritis Rheum 2005;35:1-10 1997;329:181-7 novel microemulsion systems. Eur J
2. Gerwin N, Hops C, Lucke A. 7. Gonzalez E, Cruz C, Nicolas R, et al. Pharm Biopharm 1997;43:37-42
Intra-articular drug delivery in Long-term effects of nonsteroidal 12. Dreher F, Walde P, Walter P, Wehrli E.
osteoarthritis. Adv Drug Deliv Rev anti-inflammatory drugs on the Interaction of a lecithin microemulsion
2006;58:226-42 production of cytokines and other gel with human stratum corneum and its
3. McKay N, Kelly CA. Extra-articular inflammatory mediators by blood cells of effect on transdermal transport.
features of rheumatoid arthritis. Medicine patients with osteoarthritis. J Control Release 1997;45:131-40
2006;34:383-6 Agents Actions 1994;41:171-8 13. Schmalfus U, Neubart R, Wohlrab W.
4. Zarrin Ghalam Moghaddam J, 8. Shaikh IM, Jadhav KR, Gide PS, et al. Modification of drug penetration into
Naghade M. Comparison of the effect of Topical delivery of aceclofenac from human skin using microemulsions.
Hirudo medicinals and Pyroxicam lecithin organogels: preformulation study. J Control Release 1997;46:279-85
capsule on reduction of inflammation Curr Drug Deliv 2006;3:417-27 14. Attwood D, Currie LRJ, Elworthy PH.
and pain in the primary stages of 9. Osborne DW, Ward AJ, Neil KJ. Studies of Solubilized miceller solutions.
rheumatoid arthritis of knee. Iranian J Microemulsions as topical delivery Phase studies and particle-size analysis of
Pharm Res 2004;3(Suppl):43-3 vehicles: in-vitro transdermal studies of a solution formed with non-ionic
5. Escribano E, Calpena AC, Queralt J, model hydrophilic drug. surfactants. J Colloid Interface Sci
et al. Assessment of diclofenac J Pharm Pharmacol 1991;43:450-4 1974;46:249-54
permeation with different formulations: 10. Trotta M, Pattarino F, Gasco MR. 15. Edwards DA, Langer R. A linear theory
anti-inflammatory study of a selected Influence of counter ions on the skin of transdermal transport phenomena.
formula. Eur J Pharm Sci permeation of methotrexate from J Pharm Sci 1994;83:1315-34
2003;19:203-10 water-oil microemulsions. 16. Kulkarni SK, Jain NK. Pharmacological
6. Yamazaki R, Kawai S, Mastsuzaki T. Pharm Acta Helv 1996;71:135-40 and pharmacokinetic studies on marketed
Aceclofenac blocks prostaglandin

Expert Opin. Drug Deliv. (2013) 10(4) 419


S. Choudhury et al.

gel formulations of nimesulide. 21. Eccleston J. Microemulsions. In: 25. Van-Abbe NJ, Nicholas P, Boon E.
Indian Drugs 2001;38(2):63-6 Boylan JC, Swarbrick J, editors. Exaggerated exposure in topical irritancy
17. Babota S, Alam MS, Sharma S, et al. Encyclopedia of pharmaceutical and sensitization testing. J Soc
Nanocarrier-based hydrogel of technology. Volume 9 Marcel Dekker; Cosmet Chem 1975;26:173-87
betamethasone dipropionate and salicylic NY: 1994. p. 375-421
acid for treatment of psoriasis. Int J 22. Craig DQM, Barker SA, Banning D, Affiliation
Pharm Investig 2011;1:139-47 Booth SW. An investigation into the Sandipan Dasgupta2, Sanjay Dey1,
18. Winter CA. Anti-inflammatory testing mechanisms of self-emulsification using Supratik Choudhury1 & Bhaskar Mazumder†1

methods: comparative evaluation of particle size analysis and low frequency Author for correspondence
1
dielectric spectroscopy. Int J Pharm Assistant Professor,
indomethacin and other agents.
Nonsteroidal Anti Inflammatory Drugs 1995;114:103-10 Department of Pharmaceutical Sciences,
1965;52:190-202 23. Lawrence MJ, Rees GD. Dibrugarh University, Dibrugarh-786004,
Assam, India
19. Pouton CW. Formulation of Microemulsion-based media as novel
Tel: +919435256182;
Expert Opin. Drug Deliv. Downloaded from informahealthcare.com by University Library Utrecht on 03/18/13

self-emulsifying drug delivery systems. drug delivery systems. Adv Drug


Deliv Rev 2000;45:89-121 E-mail: bhmaz@yahoo.co.in
Adv Drug Deliv Rev 1997;25:47-58 2
Research Scholar,
20. Warisnoicharoen W, Lansley AB, 24. Klang V, Matsko N, Zimmermenn A, Dibrugarh University, Department of
Lawrence MJ. Nonionic oil-in water et al. Enhancement of stability and skin
Pharmaceutical Sciences, Dibrugarh-786004,
microemulsions: the effect of oil type on permeation of sucrose stearate and Assam, India
phase behavior. Int J Pharm cyclodextrins in progesterone
2000;198:7-27 nanoemulsion. Int J Pharm
2010;393:152-60
For personal use only.

420 Expert Opin. Drug Deliv. (2013) 10(4)

You might also like