You are on page 1of 12

122 Send Orders for Reprints to reprints@benthamscience.

ae
Current Drug Delivery, 2018, 15, 122-133

RESEARCH ARTICLE
ISSN: 1567-2018
eISSN: 1875-5704

Impact
Factor:
1.478

Formulation of Niosomal Gel for Enhanced Transdermal


Lornoxicam Delivery: In-Vitro and In-Vivo Evaluation
BENTHAM
SCIENCE

Mohamed Shafik El-Ridy1, Soad Aly Yehia2, Amira Mohamed Mohsen1,*, Sally A. El-Awdan3 and
Asmaa Badawy Darwish1

1
Pharmaceutical Technology Department, National Research Centre, Dokki, Cairo, Egypt; 2Pharmaceutics
Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt; 3Pharmacology Department, National Research
Centre, Dokki, Cairo, Egypt

Abstract: Background: The objective of this study was to investigate the potential of niosomal gels as
a transdermal delivery system to improve the permeation and anti-inflammatory activity of Lornoxicam
(LX).
Methods: LX niosomes were prepared by thin film hydration technique and were characterized using
Transmission Electron Microscopy (TEM), Differential Scanning Calorimetry (DSC), Particle Size
analysis and Zeta potential determination. LX niosomal gel/LX loaded gel were prepared using Car-
bopol 934 (2%) and were evaluated for their physical appearance, pH and rheological behaviour. Ex
vivo skin permeation test was performed on dorsal region of wistar rats. In vivo studies comprised skin
ARTICLE HISTORY
irritation test and anti-inflammatory activity study.
Received: October 28, 2016
Revised: January 01, 2017 Results: The prepared LX niosomes exhibited an entrapment efficiency of more than 66% and a parti-
Accepted: February 13, 2017 cle size diameter ranging from 295 nm to 1298 nm, with negatively charged zeta potential. TEM elec-
DOI: tron micrographs revealed spherical shaped vesicles. The release pattern of drug was analyzed and
10.2174/1567201814666170224141548
found to follow Higuchi’s model. Rheology studies revealed the pseudoplastic behaviour of LX
niosomal gel. They exhibited a one and half fold increase in drug permeated through rat skin, when
compared to free drug. Skin irritation test proved the non-irritancy of LX niosomal gels, when applied
to dorsal region of Wistar rats. Percentage edema inhibition of LX niosomes was significantly higher
(P<0.05) than that of free LX group showing an enhanced anti-inflammatory activity of LX niosomes.
Conclusion: These findings revealed that LX loaded niosomal gels could be a potential transdermal
Current Drug Delivery

drug delivery system.

Keywords: Anti-inflammatory, lornoxicam, niosomal gel, niosomes, permeation, transdermal.

1. INTRODUCTION minor to moderate inflammation and pain in rheumatoid ar-


thritis and osteoarthritis [5]. Parenteral formulations that lead
Controlling inflammatory disorders should comprise a
to immediate release of LX are available for intramuscular
stepwise way to the usage of therapeutic agents. Critical
and intravenous (4 mg/ml) route of administration. The dose
goals to reduce the severity of symptoms comprises relieving
that is recommended by oral route of administration is
of pain and reduction of inflammation [1]. Non-steroidal 8-16 mg/day and the total dose/day should not exceed 16 mg
anti-inflammatory drugs (NSAIDs) are among the most
[6, 7].
widely used therapeutics, prescribed for the treatment of pain
and inflammation [2, 3]. It was stated that its analgesic activity is analogous to the
analgesic effect of opioids. The oral bioavailability of LX is
Lornoxicam is one of the non-steroidal anti-inflammatory
90-100%. It has a short half-life (3-5 hrs) [8]. The most
drugs, belonging to the class oxicam, with analgesic, antipy-
common oral dosages of LX that was used in clinical trials
retic and anti-inflammatory effect [4] .It is recommended for
were 4 mg twice or 3 times daily or 8 mg twice daily for
controlling arthritic conditions [9]. In acidic pH of the stom-
*Address correspondence to this author at the Pharmaceutical Technology ach LX has a very poor solubility [10]. Thus, it remains in
Department, National Research Centre, 33 El-Buhouth Street, Dokki,12622, contact with the stomach wall for a long period leading to
Cairo, Egypt; Tel: +202 3335456; Fax: +202 33370931;
E-mail: amiramohsen80@hotmail.com
gastric irritation and peptic ulcers [11]. In addition, the

1875-5704/18 $58.00+.00 © 2018 Bentham Science Publishers


Formulation of Niosomal Gel for Enhanced Transdermal Lornoxicam Delivery Current Drug Delivery, 2018, Vol. 15, No. 1 123

mechanism of action of LX comprises inhibition of cy- 2.2. Methods


clooxygenase isoenzymes which causes inhibition of the
An increasing number of NIS has been found to formu-
synthesis of prostaglandin from arachidonic acid. Prosta-
late niosomes among which sorbitan esters are much more
glandins have a significant role in protecting the mucosa of
common. Hao et al. [27] suggested that the length of alkyl
the gastrointestinal tract (GIT). It acts by hindering the secre-
chain is a crucial factor of permeability and that long chain
tion of gastric acid. Thus, inhibition of prostaglandin synthe- surfactants produce high drug entrapment into vesicles. Ac-
sis usually leads to GIT complications such as ulceration, cordingly, Span 40 was selected as it is one of the surfactants
bleeding and dyspepsia [12]. Although LX has better accept- of choice for the formation of the non-ionic vesicles. Choles-
ability compared to other NSAIDs, different side effects, terol was used with Span40 as an enhancer of niosomal
revealed in the GIT, have been reported for LX. Upon oral membrane rigidity [28, 29]. Charge inducing agents (CIA)
administration, renal and hematological side effects may also such as DCP (negative charge inducing agent) and SA (posi-
occur. On the other hand, parenteral administration of LX is tive charge inducing agent) were added to the niosomal bi-
not preferred due to chronic complaints [13]. layers for investigation of charge effect. The inclusion of
An alternative route to overcome the GIT side effects of charge inducing agents in niosomes tends to stabilize them
NSAIDs, including LX, can be the transdermal route of ad- [30]. Furthermore, it was reported that niosomes constructed
ministration [14, 15]. Transdermal application of drugs has from cholesterol, NIS and dicetyl phosphate were effective
many benefits over oral and intravenous routes of admini- in increasing skin permeation of frusemide across mouse
stration such as absence of first- pass metabolism and inci- skin as compared to conventional formulations [31].
dence of systemic toxicity [16]. Inflammation can be treated
using transdermal patches that also exhibit systemic effects. 2.2.1. Preparation of LX Niosomes
These systems show an improved patient compliance be- Niosomes were prepared by the thin film hydration
cause of their easy application and ability to discontinue method [24]. In a 100 ml round-bottom flask, 10 mg LX and
treatment whenever being requested [17]. accurately weighed quantities (100 mg) of Span40 and Ch
Nowadays, different nanotechnology based formulations with or without CIA were dissolved in chloroform. The or-
are available for topical application of drugs and cosmetics. ganic solvent was slowly evaporated under reduced pressure
Proniosomes [18], solid lipid nanoparticles [12], nanoemul- using the rotary evaporator (Büchi rotavapor-M/HB-140,
sion gels [19] and transdermal matrix patches [20] of LX Technik AG, Switzerland), at 56-58°C [27]. After chloro-
were previously studied and reported. Vesicular systems form evaporation, the thin film formed on the inner wall of
such as liposomes and niosomes represent an important drug the rotating flask was then hydrated with 10 ml phosphate
delivery system [11]. The advantages of niosomes over buffered saline (PBS) pH=7.4 pre-warmed to 56°C-58°C.
liposomes include enhanced chemical and physical stability Composition of different prepared LX loaded niosomes is
[21], lower cost and ease of use of surfactants [22]. listed in Table 1.
Niosomes is a vesicular system which is composed of non-
ionic surfactants (NIS). In the aqueous media, these NIS self- 2.2.2. Characterization of Prepared LX Niosomes
assemble to produce bilayer structures [23, 24]. Thus, it can 2.2.2.1. Entrapment Efficiency Percentage (E.E. %)
encapsulate both hydrophilic and hydrophobic drugs into this
bilayer structure [25]. Niosmes is a very beneficial drug de- LX niosomes was separated from the free unentrapped
livery system with several applications [22, 25, 26]. drug by cooling centrifugation at 5400 xg, at 4°C, for 30
minutes using the cooling centrifuge (Union 32R, Korea)
The objective of the present study is to develop trans- [32, 33]. The niosomal pellets were then washed with 10 ml
dermal niosomal gel of LX to improve the permeation and PBS and recentrifuged. The supernatant was filtered through
anti-inflammatory activity of LX and to enhance its patient 0.22 m Millipore filter (Millipore, USA). Then the free
compliance by providing sustained release medication. The drug content in supernatant was analyzed [11, 18] spectro-
current research work comprises an in vitro evaluation in- photometrically at max 375 nm, after comparison to a pre-
cluding preparation and characterization of LX niosomes, constructed calibration curve (n=3, linearity range: 2-20
in vitro release study as well as in vivo evaluation of the op- g/ml, R2=0.9999). Drug entrapment efficiency was calcu-
timized LX niosomal gel. lated according to the following equation:
E.E. %= [Qt – Qs]/Qt * 100 Eq. (1)
2. MATERIALS AND METHODS Where E.E. is the entrapment efficiency, Qt is amount of LX
2.1. Materials in suspension added, and Qs is amount of LX detected only
in the supernatant [17].
LX was a kind gift sample from Eva pharma Co., Cairo,
Egypt. Sorbitan monopalmitate (Span 40), Dihexadecylhy- 2.2.2.2. Transmission Electron Microscopy (TEM)
drogen-Phosphate (Dicetyl Phosphate) (DCP) and Choles-
terol (Ch) were obtained from Sigma-Aldrich Chemie GmbH, Transmission electron microscopy was performed to de-
Germany. Octadecylamine (Stearylamine) (SA) was pur- termine the morphological characteristics of vesicles [34].
chased from Sigma-Aldrich Co., St. Louis, Mo., USA. Chlo- Samples were diluted five folds with bidistilled water and
roform HPLC was obtained from Panreac Quimica SA, Bar- mixed well prior to the examination. One drop of the sample
celona, Spain. All other chemicals were of analytical grade. was placed onto 300-mesh carbon coated grid and allowed to
dry to a thin film [35]. Before complete drying of this film
124 Current Drug Delivery, 2018, Vol. 15, No. 1 El-Ridy et al.

Table 1. Composition of different LX niosomal formulations.

Molar Ratio
Formula Niosomes Type Charge Inducing Agent
Span 40 Ch
DCP SA

F1 Neutral 1 1 _ _

F2 Negative 1 1 0.1 _

F3 Positive 1 1 _ 0.1

F4 Neutral 2 1 _ _

F5 Negative 2 1 0.2 _

F6 Positive 2 1 _ 0.2

on the grid, it was negatively stained with 1% phosphotung- diluted tenfold with bidistilled water. The samples were then
estic acid. One drop of the freshly prepared stain was added transferred to a quartz cuvette and measured at room tem-
and allowed to air dry on a filter paper. After drying, the perature. Zeta potential studies were performed in triplicate.
samples were then examined by the TEM (JEOL, JEM-1230,
Tokyo, Japan.), with an accelerating voltage of 80KV [36]. 2.2.3. Preparation of LX Niosomal gel/ LX Loaded Gel
The experiment was performed at room temperature.
Preparation of LX niosomal gel of selected niosomal
2.2.2.3. Differential Scanning Calorimetry (DSC) dispersion (F1) took place by adding 200 mg of Carbopol
934 in 10 ml niosomal dispersion with continuous stirring
The thermal properties of selected LX niosomes were using a glass rod. Triethanolamine (TEA) was added drop
analyzed using DSC calibrated with indium (DSC-60 with wise for neutralization till reaching the desired skin pH (6.8-7)
TA-60 WS thermal analyzer, Shimadzu, Tokyo, Japan). [41].
Thermal analysis was also performed on plain (drug-free)
A gel base was prepared by sprinkling the weighed
niosomal formulations of the selected formulations F1 and
amount of Carbopol 934 (2% w/w) in appropriate amount of
F4. Niosomal dispersions were lyophilized before DSC
distilled water to form an aqueous dispersion [42]. This dis-
analysis using freeze dryer (VirTis Freeze Drying Equip-
persion was stirred, by magnetic stirrer, for 2 hrs. A known
ment, Snijders Scientific). Thermograms were analyzed us-
amount of LX (equivalent to 1% w/w) was added to the gel
ing Shimadzu TA-60 software. The niosomal individual
base and properly dispersed, with continuous stirring, until
components of niosomes were also analyzed. A known
homogenous gel was formed. Drops of TEA were added till
amount of the lyophilized niosomes was placed in a hermeti-
reaching the required pH. All the samples were stored in
cally sealed aluminum pan and scanned at a rate of 5°C/min
refrigerator (4°C) for at least 24 hours prior to performing
[37] over a temperature range of 20-300°C with nitrogen
rheological measurements [43].
purging (25ml/min).
2.2.4. Evaluation of Free Drug/LX Niosomal Gel
2.2.2.4. Vesicular Size Analysis
The prepared gels were evaluated via the following
Vesicular size for the prepared niosomes was determined
studies.
by dynamic light scattering method based on laser diffraction
in a multimodal mode using the Malvern particle sizing sys- 2.2.4.1. Physical Appearance
tem (Malvern, Nano Series ZS90, Malvern Instruments, Ltd.,
UK) [38, 39] with He-Ne laser at 632.8 nm at a scattering The gels were observed for their clarity and homogeneity
angle of 90.0° [40], which is capable of measuring vesicles by visual examination. They were also checked for the pres-
in the 1nm to 5 m size range. The niosomal preparation was ence of any aggregates or clumps.
diluted with bidistilled water (1:100 vol/ vol). The sample
was then transferred to a quartz cuvette and examined at 2.2.4.2. Measurement of pH
room temperature. Size distributions were displayed in term The pH of the gels was measured using digital pH meter
of number versus vesicle size. The polydispersity index (P.I.) (Thermo Scientific Orion VERSA STARTM Advanced Elec-
was determined as a measure of homogeneity [14]. trochemistry Meter, VSTAR 92, Thermo Fisher Scientific
Inc., USA) at room temperature. This study was performed
2.2.2.5. Zeta Potential Determination to assure that the pH of the developed gels are close to skin
The zeta potential was measured on the bases of laser pH, after being kept in refrigerator for 24 hrs. The determi-
diffraction analysis using Zeta sizer (Malvern, Nano Series nations were carried out in triplicate and the averages of
ZS90, Malvern Instruments, Ltd., UK). The niosomes were three readings were noted and S.D. was determined.
Formulation of Niosomal Gel for Enhanced Transdermal Lornoxicam Delivery Current Drug Delivery, 2018, Vol. 15, No. 1 125

2.2.4.3. Rheological Behavior cient (R2) were calculated from the linear curve obtained by
regression analysis of these plots.
Rheological behavior of prepared niosomal gel was
measured using parallel-plate rheometer (Anton Paar, Mt = M0 + k0t Eq. (2)
Physica MCR 301, Germany) at room temperature. The
shear stress and viscosity were determined while increasing Mt = M0 (e –k1t) Eq. (3)
shear rate progressively (from 1 to 200 sec-1) and then 1/2
Mt = kH.t Eq. (4)
gradually decreasing (from 1 to 200 sec-1) [44]. Thus
rheogram curves were constructed. The flow behavior was where Mt and M0 are amounts of drug released in the release
then analyzed by regression analysis of the log shear stress medium at time t and at t =0, respectively, k0, k1 and k H are
vs. log shear rate through following equation: release constants of the zero-order, first-order and Higuchi
square root model, respectively.
Log G = n Log F – Log  Eq. (2)
-1
Where: G is shear rate (sec ), F is shear stress (Pa),  is vis- 2.2.7. Ex Vivo Skin Permeation Experiment
cosity (Pa.s) and n (Farrow’s constant) is slope of log G
Skin permeation studies ware evaluated on hairless ab-
against log F plot, and it denotes deviation from Newtonian
dominal Wistar rat skin using Franz diffusion cell [50]. The
flow behavior. It rises as the flow becomes increasingly non-
Newtonian. When n>1, it indicates pseudoplastic flow or receptor volume was 63 ml (phosphate buffer saline pH 7.4)
[11, 12] and the surface area of the release membrane was
shear thinning and when n<1 this indicates dilatant flow or
3.14 cm2. The donor side was charged with 2 gm of the in-
shear thickening flow [45].
vestigated gel (Free drug in gel or LX niosomal gel). The
solution in the receptor side was stirred with magnetic stirrer
2.2.5. In Vitro Release Studies
bar adjusted at 500 rpm [51] and maintained at 37oC. Sam-
2.2.5.1. In Vitro Release of LX Niosomes ples (2 ml) were withdrawn at different time intervals (1, 2,
4, 6 and 24 h) and replaced by fresh medium to maintain sink
This study was conducted on neutral and negatively
condition [11]. Drug concentration was detected using a
charged niosomal dispersions. Positively charged niosomes;
validated HPLC method. HPLC Agilent 1260 infinity sys-
F3, and F6, were not included in this study and in further in
tem, equipped with MWD 1260 (Germany) was used. The
vivo studies due to their larger particle size revealed, low
system employed consisting of a quaternary pump, 1260
stability indicated by zeta potential results, in addition to
ALS degasser and variable-wavelength detector. The sam-
their aggregation observed by TEM micrographs. The ples were separated on a C18 column (Zorbax eclipse plus,
amount of LX released from different niosomal formulations
4.6 x 250 mm, Germany) with 0.1 M sodium dihydrogen-
was determined using the dialysis bag diffusion technique
phosphate buffer (pH 6)/ methanol (50:50, v/v) at a flow-rate
[46]. The dialysis bag (molecular weight cut off 12000–
of 1 ml/min. The detection absorption was at 372 nm [52].
14000) was soaked in PBS (pH 7.4) for 12h before use [12].
The linearity range of the calibration curve for LX was 10 -
The cellulose bag was filled with niosomal formulations
2000 ng/ml. The standard curve of LX in plasma was linear
equivalent to 2 mg drug. Then, the cellophane bag was im- and the regression coefficients was R2=0.9992 (n=3), indi-
mersed in a beaker containing 100 ml PBS (pH 7.4) [18, 46].
cating good linearity.
Sink conditions was maintained throughout the experiment
[47]. The beaker was placed in a water bath shaker to main- The cumulative amount of LX permeated per unit dialy-
tain the temperature at 32 ± 2ºC, with speed of 100 rpm. sis membrane area (g/cm2) was plotted versus time and the
permeation profiles were constructed. Linear regression
At predetermined time intervals (1, 2, 3, 4, 5, 6, 7, 8 and analysis was used to calculated steady state flux (Jss) of
24 hrs); samples were withdrawn for analysis and replaced
drug. The permeability co-efficient (Kp) of LX, through the
with equal volume of fresh PBS (pH 7.4) at 32 ± 2ºC to
stratum corneum, was calculated using the following equa-
maintain a constant volume. The amount of drug in the col-
tion [53]:
lected samples was detected spectrophotometrically. Each
result is the mean of at least four determinations. The per- Kp = Jss / C Eq. (5)
centage of drug release was plotted as a function of time.
Where, C is the initial concentration of the drug in the donor
2.2.5.2. In Vitro Release Study of LX Niosomal Gel compartment. The penetration enhancing effect was calculated
in terms of enhancement ratio (ER) by using the equation:
In vitro release study was conducted on selected LX
niosomes (F1) after being incorporated in gel, as well as free ER = Jss of formula/ Jss of control Eq. (6)
LX loaded gel. The experiment was performed on the same
conditions as previously mentioned in 2.2.4.1. 2.2.8. In Vivo Biological Evaluation of LX Niosomes
2.2.8.1. Animals
2.2.6. Kinetic Study of Release Profiles of LX Niosomes
and LX Niosomal Gel Albino Wistar rats weighting were used in all in vivo
studies. Animals were housed in polypropylene cages and
After completing in vitro release profiles of all the inves- were given standard diet and water. They were kept at
tigated batches, the data were treated with different mathe- 25±1ºC with at 12 hr. light /dark cycle. The in vivo experi-
matical models, i.e., zero order kinetics, first order kinetics ments were conducted in accordance with ethical procedures
[48] and Higuchi model [49], as described in Eqs. (2-4). The and policies approved by the Medical Research Ethical
release rate constant (K) values and the correlation coeffi-
126 Current Drug Delivery, 2018, Vol. 15, No. 1 El-Ridy et al.

Committee of the National Research Centre, Cairo, Egypt, 2.2.9. Data Analysis and Statistics
registration number 14078.
Data are expressed as mean ± standard deviation (S.D.).
Statistical analysis was performed using SPSS software (ver-
2.2.8.2. Skin Irritation Test
sion 22.0; IBM Co., USA). Analysis of variance (ANOVA,
The skin irritation of LX niosomal gel formulation (F1) single factor) followed by Fisher LSD’s post-hoc test, was
was evaluated in terms of biological investigation, on white employed in the statistical analysis of the determined pa-
albino Wistar rats (120-160 gm) based on the method de- rameters. A P-value < 0.05 was considered statistically
scribed by Draize et al. [54]. The rats were anesthetized with significant.
thiopental (60 mg/kg) injection (i.p) then the dorsal side of
the rat was shaved 12 hr before the beginning of the experi-
3. RESULTS AND DISCUSSION
ment. The animals were divided into five groups each group;
n=3 rats. Group I served as negative control while group II 3.1. Entrapment Efficiency Percentage (E.E. %)
acted as positive control and received 0.8% (v/v) aqueous
formalin solution as a standard irritant [55]. Group 3 re- The entrapment efficiencies (E.E. %) of all niosomal
ceived niosomal gel free from LX. Groups 4 and 5 received formulations are revealed in Table 2. The effects of several
LX niosomal gel (F1) and free LX in gel, respectively, for 3 factors including the effect of different surface charges, dif-
day. The sites of application was checked for edema and ferent molar ratios and effect of non-ionic surfactant type on
erythema after 24 hours, and then graded from zero to 4 ac- the entrapment efficiency of LX were studied.
cording to a visual scoring scale, performed by the same
investigator. The scoring followed the following manner: 0, 3.1.1. Effect of Surface Charge on LX E.E. %
none; 1, slight; 2, well defined; 3, moderate; and 4, scar or Results, tabulated in Table 2, reveal that for the molar
ulcer formation. The edema scale was: 0, none; 1, slight; 2, ratio, Span 40: Ch: CIA (1:1:0.1), neutral LX niosomes ex-
well defined; 3 moderate; and 4, severe. Photos were taken hibited the highest E.E. % (66.11 ± 2.50%) showing a sig-
for erythema visualized on some rats’ application sites. The nificant difference (P< 0.05) when compared to negatively
primary irritancy index (PII) was calculated for each group (58.72 ± 1.09%) and positively (54.5 ± 2.31%) charged
according to the equation [54]: niosomes. Same sequence was noticed for the molar ratio
Primary irritancy index (PII) = Mean of erythema + Mean of edema
Span 40: Ch: CIA (2:1:0.2). This can be attributed to the
Eq. (7) presence of higher Ch content in neutral niosomes. Choles-
terol is known to be one variant in the niosomal structure
The formulation were classified as non-irritant if (PII < 2), which could control retention of drugs by increasing the
irritant if (PII =2-5) and highly irritant if (PII=5-8). packing of surfactants molecules, reducing bilayer perme-
ability [58]. The inclusion of DCP or SA tend to decrease the
2.2.8.3. In Vivo Anti-Inflammatory Activity amount of LX entrapped and that decrease may be due to the
Anti-inflammatory activity of selected LX niosomal gel repulsion between the niosomal bilayers which led to in-
was evaluated using carrageenan induced rat paw edema crease of the leaking space in the bilayer membranes and
model. Animals were divided into five groups; n=6. Group 1 therefore permit the release of the encapsulated drug. These
acted as control. Group 2 received blank niosomal formula- results come in accordance with those reported for the incor-
tions of (F1) in gel. Group 3 received free LX in gel. Groups poration of charge inducing agents into indomethacin
4 and 5 received LX niosomal (F1) gel and market product, liposomes [59], acetazolamide liposomes [60], and gentamy-
resoectively. cin niosomes [61].
At the level of tibiotarsic articulation, a mark was made
3.1.2. Effect of Molar Ratio on E.E.%
on the right paw. The basal paw volume was measured by a
volume displacement method, previously described by The results revealed in Table 2 show that neutral, nega-
Swingle, et al. [56]; employing a water digital plethysmome- tively and positively charged LX niosomes of molar ratio
ter 7500 (Ugo Basile, Comerio, Italy). Carrageenan suspen- Span 40:Ch (1:1) and Span 40: Ch :CIA (1:1:0.1), F1, F2 and
sion (0.1 ml of 1% w/v, in saline) was injected into the sub- F3, exhibited slightly higher entrapment efficiencies than
plantar surface of the right hind paw. Following this, the those prepared using the molar ratio Span 40:Ch (2:1) and
formulations were applied on the shaved dorsal region of Span 40: Ch: CIA (2:1:0.2), respectively. These findings
animals and the paw volume was measured again at several might be attributed to the higher Ch content that reached up
time intervals (1, 2, 3, 4, 5, 6 and 24 hrs) after carrageenan to 50 mole % for the molar ratio Span 40: Ch (1:1) and Span
injection. The increase in paw volume was calculated as per- 40: Ch: CIA (1:1:0.1) compared to the molar ratio Span: Ch
centage of edema compared to the basal paw volume accord- (2:1) and Span: Ch: CIA (2:1:0.2) with a Ch content of only
ing to the following equation [57]: 30 mole %. Recently, Confalonieri et al. [62] reported that
increasing the Ch content resulted in an improvement of the
Edema inhibition (%)  Vuntreated - Vtreated X 100/Vuntreated total amount of drug encapsulated due to an increase in the
Eq. (8) lipophilic behavior and crystallinity of the surfactant bilayer
Where, Vtreated is the volume of the paw after the topical of niosomes. Accordingly, we can conclude that LX, which
treatment of gel and Vuntreated is the volume of the paw with- is a highly hydrophobic drug, intercalated in these lipophilic
out treatment portions.
Formulation of Niosomal Gel for Enhanced Transdermal Lornoxicam Delivery Current Drug Delivery, 2018, Vol. 15, No. 1 127

Table 2. Characterization of LX loaded Span 40 niosomes.

Formula Particle Size (nm) PDI Zeta Potential (mV) ± S.D. E.E. (%) ± S.D.

F1 295.3 0.391 -46.9 ± 9.83 66.11 ± 2.50

F2 531.2 0.410 -53.6 ± 6.78 58.72 ± 1.09

F3 1298 0.259 -21.2 ± 5.48 54.50 ± 2.31

F4 642.9 0.386 -56.9 ±11.6 65.68 ± 3.13

F5 740.9 0.325 -69 ± 7.4 57.62 ± 0.83

F6 955.4 0.492 -21 ± 5.31 42.83 ± 0.54

3.2. Niosomes Characterization main constituent. A clear change in the phase transition tem-
perature of Span 40 was detected with clear changes in the
3.2.1. Transmission Electron Microscopy (TEM)
enthalpy in addition to peak broadening. The DSC thermo-
Fig. (1) reveals different vesicle shapes of LX niosomal grams of LX niosomes, revealed the disappearance of the
formulations, F1-F6, at different magnification powers. The melting exothermic peak of LX and shifting of the endo-
vesicles are well identified and exhibit a sphere like shape. thermic peak of span 40. The disappearance of the character-
They also reveal well-stained niosomal vesicles, where the istic peak of drug and shifting and/or broadening of the en-
outer lipophilic domain is black stained and the inner hydro- dotherms of surfactant recommends possible contact of drug
philic domain is light stained. with bilayer components and may indicate the improved en-
trapment of LX into these vesicles [64].
3.2.2. Differential Scanning Calorimetery (DSC)
3.2.3. Vesicular Size Analysis
Fig. (2) reveal the themograms as well as the phase tran-
sition temperatures (Tc) of the free drug LX, LX loaded 3.2.3.1. Effect of CIA on Vesicle Diameter of LX Niosomes
niosomes as well as individual components of niosomes in- The results, tabulated in Table 2, reveal that charged
cluding, Span 40, Ch, DCP and SA. Thermograms of span
niosomes (F2, F3/F5, F6) showed larger vesicular sizes
40 showed sharp endothermic peak at 50.57°C, indicating its
compared to their neutral counterparts (F1 and F4), respec-
crystallinity, while the thermogram of Lornoxicam showed
tively. Incorporation of a CIA in niosomes usually leads to
sharp exothermic peak at 221.54°C, which is in accordance
an improvement in the spacing between adjacent bilayers,
with its reported melting point [63].
leading to an increase in the size of the internal aqueous
DSC thermograms of plain niosomal formulations re- compartment which causes the formation of larger niosomes
vealed a change in the phase transition temperature of the [65].

200 nm 500 nm 1 mm

F1 F2 F3

1 mm 1 mm 2 mm

F4 F5 F6

Fig. (1). Transmission electron microscopy micrographs of LX loaded niosomes.


128 Current Drug Delivery, 2018, Vol. 15, No. 1 El-Ridy et al.

0 50 100 150 200 250 300 respectively). The negative values of the zeta potential for
0.0 neutral niosomes might be attributed to the adsorption of
-0.3
-0.6 F1 LX Niosomes
counter ions or the preferential adsorption of hydroxyl ions
-0.9 at the vesicle surface [30].
0.00
-0.33 Negatively charged LX niosomes exhibited higher nega-
-0.66 F1 drug free niosomes
tive ZP values, i.e., -53.6 ± 6.78 mV and -69 ± 7.4mV for F2
-0.99
18.3
and F5, respectively, showing higher stability. Positively
12.2 Lornoxicam
charged LX niosomes showed lower ZP values (-21.2 ± 5.48
6.1 mV and -21 ± 5.31 mV) which indicates lower stability. Ac-
Endothermic

0.0
0.0
cordingly, positively charged niosomes were not included in
-6.3 further studies.
-12.6 Stearylamine
-18.9
0.0 3.3. Evaluation of Free Drug/LX Niosomal Gel
-4.1
-8.2 3.3.1. Physical Appearance
-12.3 Dicetyl Phosphate
0.0 The developed gels appeared clear and smooth. All gels
-2.9
examined were homogenous, showing no aggregates or
-5.8
-8.7 Cholesterol
clumps.
0.0
-5.6
3.3.2. pH
-11.2 S pa n 4 0
-16.8 The pH measured for the prepared LX niosomal gel/free
0 50 100 150 200 250 300 drug gel are listed in Table 3. Their pH ranged from 6.92-
Temperature (C) 6.96, showing similar pH as that of skin.

Fig. (2). Differential Scanning Calorimetry thermograms of drug, 3.3.3. Rheology Behavior
niosomal components, LX loaded niosomes and drug free niosomes.
Fig. (3) illustrates the flow curve (shear stress vs. shear
rate) and viscosity curve (viscosity vs. shear rate) of LX
3.2.3.2. Effect of Molar Ratio on Vesicular Diameter of LX niosomal gel as well as that of LX loaded gel. The upward
Niosomes and downward flow curves nearly overlapped, showing a
From the results, it can be concluded that negative LX very small gap between each other, indicating that the devel-
niosomes of the molar ratio Span 40: Ch: CIA (1:1:0.1), ex- oped gels exhibited shear thinning (pseudoplastic) behavior
hibited smaller vesicle sizes compared to their counterparts [71]. This shear thinning behavior can be ascribed to the
of the molar ratio, Span 40: Ch: CIA (2:1:0.2). Incorporation instant arrangement of the colloidal network structure into
of DCP, which is an anionic surfactant in hydrophilic nature, layers that can flow over each other more easily in the direc-
into the niosome membrane leads to water efflux into the tion of shear, thus viscosity is decreased with the increase of
bilayer and promotes separation between bilayers [66]. Thus, shear rate. Farrow’s constants (n), listed in Table 3, were
particle size and membrane thickness also increase. 3.21 and 3.90 for LX niosomal gel and free drug loaded gel,
respectively, confirming the pseudoplastic or shear thinning
On the contrary, in the case of positive niosomes of the
behavior (since n>1) [45]. These findings reveal that the de-
molar ratio Span 40: Ch: SA (1:1:0.1), containing higher
veloped systems acquired all the requirements needed for
cholesterol content, exhibited larger vesicle size than posi-
topical application.
tive niosomes of the molar ratio, Span 40: Ch: SA (2:1:0.2).
This can be attributed to the fact that cholesterol increases
3.4. In Vitro Release Profile of LX Niosomes and LX
the width of lipid bilayers and consequently increases the
Niosomal Gel
vesicle size [67].
The release profiles of drug from the investigated nioso-
3.2.3.3. Polydispersity Index (PDI) mal dispersions are demonstrated in Fig. (4). The results
reveal that the release of LX from niosomes was biphasic.
PDI values that are (< 0.3) indicate a homogenous popu-
There was an initial rapid drug release where about 11-33 %
lation, while higher PDI values moves towards heterogeneity
of the encapsulated drug was released, from various nioso-
[68, 69] . PDI values, tabulated in Table 2 calculated for LX
mal formulations as well as niosomes in gel, in the first hour.
niosomal dispersions ranged from 0.259 to 0.492, indicating
This can be explained by the fact that the lipophilic drug LX
an intermediate homogeneity.
is mostly encapsulated between the fatty acid chains of the
lipid bilayers of niosomal vesicles. This caused rapid release
3.2.4. Zeta Potential (ZP)
of drug on dispersing vesicles in buffer till reaching equilib-
The zeta potential is a measurement of stability of rium [72]. During the following 5 h, further 15-29% of LX
niosomes. It was reported that if zeta potential values are was released from various niosomal preparations. After that,
lower than (-30 mV) or higher than (+30 mV) the formula- the entrapped drug showed a sustained release profile. Dif-
tion is considered to be stable [70]. Results, revealed in ferences in the in vitro release patterns could be due other
Table 2, show that neutral LX niosomes showed negative ZP factors such as lamellarity, vesicle size and membrane fluid-
values (-46.9 ± 9.83 mV and -56.9 ±11.6 mV, for F1 and F4, ity which depends on chain length of surfactant [73].
Formulation of Niosomal Gel for Enhanced Transdermal Lornoxicam Delivery Current Drug Delivery, 2018, Vol. 15, No. 1 129

LX niosomal gel LX loaded gel


Up Up
600 1200 Down
Down

Shear stress (Pa)


500 1000

Shear stress (Pa)


400 800

300 600

200 400

100 200

0 0
0 50 100 150 200 0 50 100 150 200
Shear rate (1/s) Shear rate (1/s)

50 60

40 50
Viscocity (Pa.s)

Viscocity (Pa.s)
40
30
30
20
20
10
10

0 0
0 50 100 150 200 0 50 100 150 200
Shear rate (1/s) Shear rate (1/s)

Fig. (3). Rheograms of LX niosomal gels and LX loaded gel presenting shear stress vs. shear rate and viscosity vs. shear rate plots.

120 F1 niosomal
gel
100 Free LX gel

F1
80
Drug released (%)

F2
60
F4
40
F5
20
Free LX

0
0 5 10 15 20 25 30
Time (hrs)

Fig. (4). In vitro release profiles of different LX loaded niosomes and LX niosomal gel.

Table 3. pH and rheological behavior of LX niosomal gel and LX loaded gel.

Viscosity at Minimum Viscosity at Maximum


Formulation pH ( mean ± SD) Farrow's Constant
Shear Rate (Pa.s) Shear Rate (Pa.s)

LX niosomal gel 6.96 ± 0.11 47.3 2.86 3.21


LX loaded gel 6.92 ± 0.12 60 5.83 3.90

Fig. (4) also reveals the release pattern of the selected LX correlation coefficients (R2) values, showed that the release
niosomal gel (F1). It can be observed that the drug release of LX from niosomal formulations/niosomal gel is best-fitted
from niosomal gel occurred in two phases, a controlled re- to Higuchi’s model (diffusion mechanism), where it showed
lease phase, characterized by a relatively moderate drug re- the highest (R2) values. The results point to sustained release
lease rate, followed by a steady phase with a reduced and characteristics with a diffusion pattern of drug release, where
slow release rate which was maintained for 24 hours. niosomes act as a reservoir system for continuous delivery of
drug. These controlled release patterns of entrapped drug
3.5. Kinetics Studies of the Release Profiles reflect the high stability of these niosomal formulations [74].
Different mathematical models were employed to predict These findings confirmed that the drug was released from
the release mechanisms and compare the release profiles. niosomes by a diffusion-controlled manner, and they are in
Linear regression analysis of the mathematical models was agreement with results obtained and reported by previous
employed for the release data of LX from niosomes. The research studies [74-77].
130 Current Drug Delivery, 2018, Vol. 15, No. 1 El-Ridy et al.

35

30

Drug permeated (%)


25

20

15

10 Niosomal
gel F1
5 Free LX gel

0
0 5 10 15 20 25 30
Time (hr)
Fig. (5). Permeation profiles of LX loaded niosomal gel and LX gel.

(a)Erythema=0, Edema=0, PII=0.00 (b)Erythema=3, Edema=2, PII=5.0 (c)Erythema=1, Edema=0, PII=1.0

(d)Erythema=0, Edema=0, PII=0.0 (e)Erythema=0, Edema=0, PII=0.0 (f)Erythema=0, Edema=0, PII=0.0

Fig. (6). In vivo skin irritation study showing photos of the dorsal region of wistar rats after application of (a) control; (b) positive control; (c)
drug free niosomal gel; (d, e) optimized LX niosomal gel (F1) (f) plain LX gel.

3.6. Ex Vivo Skin Permeation acteristics enhance hydration of the skin and drug deposition
as well [61, 78].
Fig. (5) show the permeation profiles of drug from plain
gel as well as from F1 niosomal gel. Results show superior- 3.7. In Vivo Evaluation of LX Niosomes
ity of permeation of LX encapsulated in niosomes (F1) over
plain LX gel. Table 4 reveals the permeability parameters; 3.7.1. Skin Irritation Test
steady-state flux (Jss), permeability coefficient (Kp), and en-
Fig. (6) reveals the photographs taken after application of
hancement ratio (ER). There was a significant (P<0.05) im- different formulations to Carrageenan injected wistar rats.
provement in the permeation coefficient (Kp) of LX when The results show that negative control group didn’t show any
encapsulated in niosomes, 2.41x10-3 ± 9.52x10-5 cm/hr for erythema or edema. Positive control group (Fig. 5b) showed
F1niosomal gel, compared to free LX gel (1.48x10-3 ± redness, erythema and edema and scored 3 for erythrma and
12.72x10-5 cm/hr). There was about one and a half fold in- 2 for edema yielding a PII score of 5. Blank niosomal gel
crease in deposition of drug when incorporated into the showed some erythema, without any edema and PII was cal-
niosomal gel (F1), when compared to free drug in gel, as culated to be 1. Rats which received either LX niosomal gel
revealed by Er (1.62 ± 0.23). Improved skin permeation of F1 (Fig. 5d,e) or free LX gel (Fig. 5f) were free of any irrita-
LX from niosomal gel can be justified by the fact that non- tion and there were no signs of erythema or edema
ionic surfactant (Span 40) in the niosomal formulation act as (PII=0).According to Draize et al. [54], the investigated
permeation enhancers. The surfactants in vesicular form re- niosomal gels were considered to be non-irritant [PII< 2].
duce the crystallinity of the intracellular lipid bilayers of the The primary irritation indices emphasized the non-irritancy
skin and thus improve drug permeation [11]. Furthermore, of the niosomal formulation components and showed that the
the niosomal vesicles in nanometer size range show higher investigated formula is safe to be applied on the skin for the
occlusive characteristics over plain aqueous gel. These char- intended period of time.
Formulation of Niosomal Gel for Enhanced Transdermal Lornoxicam Delivery Current Drug Delivery, 2018, Vol. 15, No. 1 131

Table 4. Comparison of LX loaded niosomal gel and free LX gel in terms of steady flux (ug/cm2/hr), permeation co-efficient (cm/hr)
and enhancement ratio from ex vivo skin permeation studies, n=3.

Formulation Jss ± S.D.* (ug/cm2 /hr) Kp ± S.D.* (cm/hr) Enhancement Ratio (Er)

F1 4.83 ± 0.19 2.41x10-3 ± 9.52x10-5 1.62 ± 0.23


Free LX 3.12 ± 0.31 1.48x10-3 ± 12.72x10-5 __
*: data are significantly different from one another at P<0.05.

Plain niosomal gel Sp40: Ch (1:1)


LX gel
Niosomal gel Sp40: Ch (1:1)
Market product
30

a, b
a, b
25

a, b
Edema inhibition (%)

a, b

a, b
a, b

a
a, b
20

a, c
a

a, c

a, c, d
15
a, c
a,c
a
a

b, c, d
b, c, d

b, c, d
b, c, d

10

b, c, d
c
c

0
0 1 2 3 4 5 6 7 8
Time (hr)
Fig. (7). Paw edema inhibition percentages after application of plain niosomal gel, LX gel, LX niosomal gel and market product to car-
rageenan induced rat paw. “a” significantly different from plain niosomal gel group, “b” significantly different from LX gel group, “c” sig-
nificantly different from LX loaded niosomal gel group, “d” significantly different from market product, at P<0.05.

3.7.2. In Vivo Anti-Inflammatory Activity revealed that the release of LX from niosomes is best-fitted to
Higuchi’s model. Skin permeation studies proved improved
The percentage edema inhibition is shown in Fig. (7).
skin permeation of LX from niosomal gel. Skin irritation test
The results reveal that the percentage edema inhibition for
proved the non-irritancy of the applied niosomal gel compo-
F1 LX niosomal gel was significantly higher (P<0.05) than nents. Paw Edema study revealed enhanced anti-inflammatory
free LX group, starting 3 hours and throughout the whole
activity of LX niosomal formulations. These findings reveal
study time till 24 hours (25.5±4.32% for F1 compared to
the potential for LX encapsulation in niosomes.
11.79 ± 6.97 for free LX group). Since LX niosomal gel F1
remained superior to the free LX in its ability to suppress ETHICS APPROVAL AND CONSENT TO PARTICI-
edema, this indicates improved anti-inflammatory activity of PATE
LX niosomal system. Statistical analysis, employing one
Animal procedures were conducted in accordance with
way ANOVA followed by LSD, revealed insignificant dif-
the Medical Research Ethics Committee of the National Re-
ference between LX niosomal systems and market product.
search Centre, Cairo, Egypt, registration number 14078.
CONCLUSION HUMAN AND ANIMAL RIGHTS
LX loaded niosomes were successfully prepared yielding No Humans were used for studies that are base of this
a high drug E.E.%. The vesicles prepared were in the research.
nanometric range and exhibited a sphere like shape. DSC The reported experiments in accordance with the stan-
thermogram indicated possible contact of drug with bilayer dards set forth in the 8th Edition of Guide for the Care and
components. Rheological studies revealed that LX niosomal Use of Laboratory Animals (http:// grants.nih.gov/grants/
gel acquired pseudoplastic behavior. The release pattern of olaw/Guide-for-the-care-and-use-of-laboratory-animals.pdf)
LX from the niosomal formulations as well as niosomal gel published by the National Academy of Sciences, The Na-
was biphasic, with an initial rapid drug release that was fol- tional Academies Press, Washington DC, United States of
lowed by a slower one. The correlation coefficients values America.
132 Current Drug Delivery, 2018, Vol. 15, No. 1 El-Ridy et al.

CONSENT FOR PUBLICATION [18] Chavan, P.; Jain, B.; Jain, P. Proniosomes as drug carrier system
for transdermal delivery of lornoxicam. World J. Pharm. Pharm.
Not applicable. Sci., 2012, 1(1), 393-404.
[19] Dasgupta, S.; Ghosh, S.K.; Ray, S.; Kaurav, S.S.; Mazumder, B. In
vitro & in vivo studies on lornoxicam loaded nanoemulsion gels for
CONFLICT OF INTEREST topical application. Curr. Drug Deliv., 2014, 11(1), 132-138.
The authors declare no conflict of interest, financial or [20] Patel, B.B.; Shah, C.N. Formulation and in vitro characterization of
lornoxicam transdermal matrix patch. Int. J. Adv. Pharm., 2016,
otherwise. 5(2), 30-38.
[21] Vora, B.; Khopade, A.J.; Jain, N. Proniosome based transdermal
ACKNOWLEDGEMENTS delivery of levonorgestrel for effective contraception. J. Control.
Rel., 1998, 54(2), 149-165.
The authors express their gratitude to the Project’s Sector [22] Manconi, M.; Sinico, C.; Valenti, D.; Lai, F.; Fadda, A.M.
at the National Research Centre, Cairo, Egypt, for funding Niosomes as carriers for tretinoin, III. A study into the in vitro cu-
this work through the research group project fund number taneous delivery of vesicle-incorporated tretinoin. Internat. J.
Pharm., 2006, 311(1), 11-19.
10070115. [23] Baillie, A.F.A.; Hume, L.; Muirhead, G.; Rogerson, A. The prepa-
ration and properties of niosomes-non-ionic surfactant vesicles. J.
REFERENCES Pharm. Pharmacol., 1985, 37(12), 863-868.
[24] Azmin, M.N.F.A.; Handjani-Vila, R.M.; Stuart, J.F.B.; Vanlerber-
[1] Foye, W.O.; Lemke, T.L.; Williams, D.A. Foye's Principles of ghe, G.; Whittaker, J.S. The effect of non-ionic surfactant vesicle
Medicinal Chemistry, Lippincott Williams & Wilkins, 2008. (niosome) entrapment on the absorption and distribution of
[2] Lim, H.W.; Thorbecke, G.J.; Baer, R.L.; Gigli, I. Effect of indo- methotrexate in mice. J. Pharm. Pharmacol., 1985, 37(4), 237-242.
methacin on alteration of ATPase-positive Langerhans cell density [25] Junginger, H.; Hofland, H.; Bouwstra, J. Liposomes and niosomes,
and cutaneous sunburn reaction induced by ultraviolet-B radiation. interactions with human skin. Cosmetics Toiletries, 1991, 106(8),
J. Invest. Dermatol., 1983, 81(5), 455-458. 45-50.
[3] Bissett, D.; Chatterjee, R.; Hannon, D. Photoprotective effect of [26] HandjaniVila, R.; Ribier, A.; Rondot, B.; Vanlerberghie, G. Dis-
topical anti-inflammatory agents against ultraviolet radiation- persions of lamellar phases of nonionic lipids in cosmetic prod-
induced chronic skin damage in the hairless mouse. Photodermatol. ucts. Int. J. Cosmetic Sci., 1979, 1(5), 303-314.
Photoimmunol. Photomed., 1990, 7(4), 153-158. [27] Hao, Y.; Zhao, F.; Li, N.; Yang, T.; Li, K. Studies on a high encap-
[4] Homdrum, E.-M.; Likar, R.; Nell, G. Xefo® rapid, A novel effec- sulation of colchicine by a noisome system. Int. J. Pharm., 2002,
tive tool for pain treatment. Eur. Surg., 2006, 38(5), 342-352. 244, 73-80.
[5] Staunstrup, H.O.J.; Larsen, U.T.; Elbaek, K.; Larsen, U.; Kroner, [28] Manosroi, A.; Wongtrakul, P.; Manosroi, J.; Sakai, H.; Sugawara,
K. Efficacy and tolerability of lornoxicam versus tramadol in post- F.; Yuasa, M.; Abe, M. Characterization of vesicles prepared with
operative pain. J. Clin. Pharmacol., 1999, 39(8), 834-841. various non-ionic surfactants mixed with cholesterol. Colloids Sur-
[6] Hamza, Y.E.S.; Aburahma, M.H. Innovation of novel sustained faces B, Biointerf., 2003, 30(1), 129-138.
release compression-coated tablets for lornoxicam, formulation and [29] Foye, W.O.; Thomas, L.L.; David, A.W. Principles of Medicinal
in vitro investigations. Drug Dev. Ind. Pharm., 2010, 36(3), 337- Chemistry. Williams and Wilkins, USA. 1995, 4th edn., 335.
349. [30] Junyaprasert, V.B.; Teeranachaideekul, V.; Supaperm, T. Effect of
[7] Skjodt, N.M.; Davies, N.M. Clinical pharmacokinetics of Iornoxi- charged and non-ionic membrane additives on physicochemical
cam. A short half-life oxicam. Clin. Pharmacokinet., 1998, 34, properties and stability of niosomes. AAPS Pharm. Sci. Tech.,
421-428. 2008, 9(3), 851-859.
[8] Karna, N. Design, development and evaluation of novel sustained [31] Azeem, A.; Ahmad, F.J.; Khan, Z.I.; Talegaonkar, S. Nonionic
release bi-layer tablets of lornoxicam based on the combination of surfactant vesicles as a carrier for transdermal delivery of
hydrophilic matrix formers and basic pH modifiers. Int. J. Pharm. frusemide. J. Disp. Sci. Technol., 2008, 29(5), 723-730.
Biol. Sci., 2012, 3, 392-402. [32] El-Ridy, M.S.B.A.A.; Safar, M.M.; Mohsen, A.M. Niosomes as a
[9] Godara, S.; Srivastava, R.; Godara, R.; Bhutani, G. Lornoxicam, a novel pharmaceutical formulation encapsulating the hepatoprotec-
review of its therapeutic potential in different clinical studies. J. tive drug silymarin. Int. J. Pharmacy Pharm. Sci., 2012, 4(1), 549-
Drug Deliv. Therap., 2013, 3(2), 145-148. 559.
[10] Skjodt, N.M.; Davies, N.M. Clinical pharmacokinetics of lornoxi- [33] Kuntsche, J.; Freisleben, I.; Steiniger, F.; Fahr, A. Temoporfin-
cam. Clin. Pharmacokinet., 1998, 34(6), 421-448. loaded liposomes, physicochemical characterization. Eur. J. Phar-
[11] Deepak, P.W.K.; Vavia, P. Niosomal gel of lornoxicam for topical maceut. Sci., 2010, 40(4), 305-315.
delivery: In vitro assessment and pharmacodynamic activity. AAPS [34] Omr,i A.A.M.R. Comparison of the bactericidal action of ami-
Pharm. Sci. Tech., 2013, 14(3), 1072-1082. kacin,netilmicin and tobramycin in free and liposomal formulation
[12] Gönüllü, Ü.; Üner, M.; Yener, G.; Karaman, E.F.; Aydomu, Z. against Pseudomonas aeruginosa. Chemotherapy, 1996 b, 42, 170-
Formulation and characterization of solid lipid nanoparticles, 176.
nanostructured lipid carriers and nanoemulsion of lornoxicam for [35] Scherphof, G.L.; Velinova, M.; Kamps, J.; Donga, J.; van der
transdermal delivery. Acta Pharmaceutica., 2015, 65(1), 1-13. Want, H.; Kuipers, F. Modulation of pharmacokinetic behavior of
[13] Lin, S.-Z.; Wouessidjewe, D.; Poelman, M.-C.; Duchene, D. In liposomes. Adv. Drug Deliv. Rev., 1997, 24(2), 179-191.
vivo evaluation of indomethacin/cyclodextrin complexes gastroin- [36] Omri, A.A.M.R. Preparation, properties and the effects of ami-
testinal tolerance and dermal anti-inflammatory activity. Int. J. kacin, netilmicin and tobramycin in free and liposomal formula-
Pharm., 1994, 106(1), 63-67. tions on Gram-negative and Gram-positive bacteria. Int. J. Antimi-
[14] Yener, G.; Üner, M.; Gönüllü, Ü.; Yildirim, S.; Kiliç, P, Aslan, crob. Agents, 1996, 7, 9-14.
S.S.; Barla, A. Design of meloxicam and lornoxicam transdermal [37] Beaulac, C.; Clement-Major, S.; Hawari, J.; Lagace, J. In vitro
patches, Preparation, physical characterization, ex vivo and in vivo kinetics of drug release and pulmonary retention of microencapsu-
studies. Chem. Pharm. Bull., 2010, 58(11), 1466-1473. lated antibiotic in liposomal formulations in relation to the lipid
[15] Kavitha, K.; Rajendra, M.M. Design and evaluation of transdermal composition. J. Microencapsulation, 1997, 14, 335-348.
films of lornoxicam. Int. J. Pharma Biosci., 2011, 2(2), 54-62. [38] Halwani, M.; Mugabe, C.; Azghani, A.O.; Lafrenie, R.M.; Kumar,
[16] Khadka, D.; Ahmed, M.G.; Acharya, A. Formulation and evalua- A.; Omri, A. Bactericidal efficacy of liposomal aminoglycosides
tion of transdermal gel of lornoxicam and comparative diffusion against Burkholderia cenocepacia. J. Antimicrob. Chemothera.,
study by iontophoresis and chemical enhancers. IJAPR., 2015, 6(2), 2007, 60(4), 760-769.
40-49. [39] Mugabe, C.; Azghani, A.O.; Omri, A. Preparation and characteriza-
[17] Williams, A. Transdermal and topical drug delivery, from theory to tion of dehydration–rehydration vesicles loaded with aminoglyco-
clinical practice, Pharmaceutical Press London, 2003. side and macrolide antibiotics. Int. J. Pharmaceut., 2006, 307(2),
244-250.
Formulation of Niosomal Gel for Enhanced Transdermal Lornoxicam Delivery Current Drug Delivery, 2018, Vol. 15, No. 1 133

[40] Shekunov, B.Y.; Chattopadhyay, P.; Tong, H.H.; Chow, A.H. [60] Hathout, R.M.; Mansour, S.; Mortada, N.D.; Guinedi, A.S.
Particle size analysis in pharmaceutics, principles, methods and ap- Liposomes as an ocular delivery system for acetazolamide, in vitro
plications. Pharmaceut. Res., 2007, 24(2), 203-227. and in vivo studies. AAPS Pharm. Sci. Tech., 2007, 8(1), E1-E12.
[41] Das, S.; Haldar, P.K.; Pramanik, G. Formulation and evaluation of [61] Abdelbary, G.; El-gendy, N. Niosome-encapsulated gentamicin for
herbal gel containing Clerodendron infortunatum leaves extract. ophthalmic controlled delivery. AAPS Pharm. Sci. Tech., 2008,
Int. J. Pharm. Tech. Res., 2011, 1(3), 140-143. 9(3), 740-747.
[42] Fathalla, D.; Abdel-Mageed, A.; Abdel-Hamid, F.; Ahmed, M. In [62] Confalonieri, E.O.; Soraci, A.L.; Becaluba, M.; Denzoin, L.; Rod-
vitro and in vivo evaluation of niosomal gel containing aceclofenac riguez, E.; Riccio, B.; Tapia, O. The disposition of free and
for sustained drug delivery. Int. J. Pharm. Sci. Res., 2014. niosomally encapsulated Rac-flurbiprofen in dairy bovines. J. Vet-
http://dx.doi.org/10.15344/2394-1502/2014/105 erin. Pharmacol. Therap., 2010, 33(1), 9-14.
[43] Islam, M.T.; Rodriguez-Hornedo, N.; Ciotti, S.; Ackermann, C. [63] Ahmed, M.O.; Al-Badr, A.A. Lornoxicam. Profiles Drug Subst.,
Rheological characterization of topical carbomer gels neutralized to Excip. Relat. Methodol., 2011, 36, 205-239.
different pH. Pharmaceut. Res., 2004, 21(7), 1192-1199. [64] ElMeshad, A.N.; Mohsen, A.M. Enhanced corneal permeation and
[44] Basha, M.; Abd El-Alim, S.H.; Kassem, A.A.; El Awdan, S.; antimycotic activity of itraconazole against Candida albicans via a
Awad, G. Benzocaine loaded solid lipid nanoparticles, Formulation novel nanosystem vesicle. Drug Deliv., 2016, 23(7), 2115-2123.
design, in vitro and in vivo evaluation of local anesthetic effect. [65] Vyas, S.; Singh, R.; Asati, R. Liposomally encapsulated diclofenac
Curr. Drug Deliv., 2015, 12(6), 680-692. for sonophoresis induced systemic delivery. J. Microencapsulation,
[45] Dhawan, B.; Aggarwal, G.; Harikumar, S. Enhanced transdermal 1995, 12(2), 149-154.
permeability of piroxicam through novel nanoemulgel formulation. [66] Carafa, M.; Santucci, E.; Alhaique, F.; Coviello, T.; Murtas, E.;
Int. J. Pharm. Invest., 2014, 4(2), 65-76. Riccieri, F.; Lucaniab, G.; Torrisib, M.R. Preparation and proper-
[46] Dubey, A.; Prabhu, P. Development and evaluation of lornoxicam ties of new unilamellar non-ionic/ionic surfactant vesicles. Int. J.
loaded maltodextrin based proniosomes. Development, 2013, 5(3), Pharm., 1998, 160(1), 51-59.
865-872. [67] McIntosh, T. The effect of cholesterol content on the structure of
[47] Bhaskaran, S.; Lakshmi, P. Comparative evaluation of niosome phosphatidylcholine bilayers. Biochim. Biophys. Acta, 1978, 51,
formulations prepared by different techniques. Acta. Pharm. Sci., 43-58.
2009, 51(1), 27-32. [68] entjurc, M.; Vrhovnik, K.; Kristl, J. Liposomes as a topical deliv-
[48] Najib, N.; Suleiman, M. The kinetics of drug release from ethylcel- ery system, the role of size on transport studied by the EPR imag-
lulose solid dispersions. Drug Dev. Ind. Pharm., 1985, 11(12), ing method. J. Control. Release, 1999, 59(1), 87-97.
2169-2181. [69] Centis, V.; Vermette, P. Physico-chemical properties and cytotox-
[49] Higuchi, T. Mechanism of sustainedaction medication. Theoretical icity assessment of PEG-modified liposomes containing human
analysis of rate of release of solid drugs dispersed in solid matrices. hemoglobin. Colloids Surf. B, Biointerf., 2008, 65(2), 239-246.
J. Pharmaceut. Sci., 1963, 52(12), 1145-1149. [70] Hanaor, D.A.H.; Michelazzi, M.; Leonelli, C.; Sorrell, C.C. The
[50] Jain, S.; Chourasia, M.; Masuriha, R.; Soni, V.; Jain, A.; Jain, N.K.; effects of carboxylic acids on the aqueous dispersion and electro-
Gupta, Y. Solid lipid nanoparticles bearing flurbiprofen for trans- phoretic deposition of ZrO2 . J. Eur. Ceramic Soc., 2012, 32(1),
dermal delivery. Drug Deliv., 2005, 12(4), 207-215. 235-244.
[51] Yousuf, M.; Ahmad, M.; Usman, M.; Ali, I. Ketotifen fumarate and [71] El-Say, K.M.; Abd-Allah, F.I.; Lila, A.E.; Hassan, A.E.-S.A.;
salbutamol sulphate combined transdermal patch formulations, in Kassem, A.E.A. Diacerein niosomal gel for topical delivery, devel-
vitro release and ex vivo permeation studies. Ind. J. Pharm. Sci., opment, in vitro and in vivo assessment. J. Liposome Res., 2016,
2013, 75(5), 569. 26(1), 57-68.
[52] Radhofer-Welte, S.; Dittrich, P. Determination of the novel non- [72] Mokhtar, M.; Sammour, O.A.; Hammad, M.A.; Megrab, N.A.
steroidal anti-inflammatory drug lornoxicam and its main metabo- Effect of some formulation parameters on flurbiprofen encapsula-
lite in plasma and synovial fluid. J. Chromatography B, Biomed. tion and release rates of niosomes prepared from proniosomes. Int.
Sci. Appl., 1998, 707(1), 151-159. J. Pharm., 2008, 361(1), 104-111.
[53] Edwards, D.A.; Langer, R. A linear theory of transdermal transport [73] Weiner, N.; Williams, N.; Birch, G.; Ramachandran, C.; Shipman,
phenomena. J. Pharm. Sci., 1994, 83(9), 1315-1334. C.; Flynn, G. Topical delivery of liposomally encapsulated inter-
[54] Draize, J.H.; Woodard, G.; Calvery, H.O. Methods for the study of feron evaluated in a cutaneous herpes guinea pig model. Antimi-
irritation and toxicity of substances applied topically to the skin and crobial Agents Chemother., 1989, 33(8), 1217-1221.
mucous membranes. J. Pharmacol. Exper. Therap., 1944, 82(3), [74] Attia, I.A.; El-Gizawy, S.A.; Fouda, M.A.; Donia, A.M. Influence
377-390. of a niosomal formulation on the oral bioavailability of acyclovir in
[55] Mutalik, S.; Udupa, N. Pharmacological evaluation of membrane- rabbits. AAPS Pharm. Sci. Tech., 2007, 8(4), E106.
moderated transdermal system of glipizide. Clin. Exp. Pharmacol. [75] Hashim, F.; El-Ridy, M.; Nasr, M.; Abdallah, Y. Preparation and
Physiol., 2006, 33(12), 17-26. characterization of niosomes containing ribavirin for liver target-
[56] Swingle, K.F.; Grant, T.J.; Jaques, L.W.; Kvam, D.C. Interactions ing. Drug Deliv., 2010, 17(5), 282-287.
of anti-inflammatory drugs in carrageenan-induced foot edema of [76] Bayindir, Z.S.; Yuksel, N. Characterization of niosomes prepared
the rat. J. Pharmacol. Exp. Ther., 1970, 172(2), 423-425. with various nonionic surfactants for paclitaxel oral delivery. J.
[57] Charoo, N.A.; Shamsher, A.A.A.; Kohli, K.; Pillai, K.; Rahman, Z. Pharm. Sci., 2010, 99(4), 2049-2060.
Improvement in bioavailability of transdermally applied flurbipro- [77] Guinedi, A.S.; Mortada, N.D.; Mansour, S.; Hathout, R.M. Prepa-
fen using tulsi (Ocimum sanctum) and turpentine oil. Colloids Surf. ration and evaluation of reverse-phase evaporation and multilamel-
B, Biointerf., 2008, 65(2), 300-307. lar niosomes as ophthalmic carriers of acetazolamide. Int. J.
[58] Abd-Elbary, A.; El-Laithy, H.; Tadros, M. Sucrose stearate-based Pharm., 2005, 306(1-2), 71-82.
proniosome-derived niosomes for the nebulisable delivery of cro- [78] Ruckmani, K.; Sankar, V. Formulation and optimization of zi-
molyn sodium. Int. J. Pharm., 2008, 357(1), 189-198. dovudine niosomes. AAPS Pharm. Sci. Tech., 2010, 11(3), 1119-
[59] Srinath, P.; Vyas, S.; Diwan, P.V. Preparation and pharmacody- 1127.
namic evaluation of liposomes of indomethacin. Drug Dev. Ind.
Pharm., 2000, 26(3), 313-321.

You might also like