You are on page 1of 12

Biomedicine & Pharmacotherapy 93 (2017) 255–266

Available online at

ScienceDirect
www.sciencedirect.com

Formulation and optimization of lacidipine loaded niosomal gel for


transdermal delivery: In-vitro characterization and in-vivo activity
Mohd Qumbara , Ameeduzzafarb , Syed Sarim Imamc , Javed Alia , Javed Ahmadd ,
Asgar Alia,*
a
Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
b
Departments of Pharmaceutics, College of Pharmacy, Al-Jouf University, Al-Jouf, Saudi Arabia
c
Departments of Pharmaceutics, Glocal School of Pharmacy, Glocal University, Saharanpur, UP, India
d
Departments of Pharmaceutics, College of Pharmacy, Najran University, Najran, Saudi Arabia

A R T I C L E I N F O A B S T R A C T

Article history:
Received 15 March 2017 The aim of the present research work is to formulate lacidipine (LAC) loaded niosomes formulation for
Received in revised form 29 May 2017 the management of hypertension by thin film hydration technique. The developed formulations were
Accepted 13 June 2017 statistically optimized by four factors, three levels Box–Behnken design and were evaluated for vesicle
size, entrapment efficiency, and flux. The optimized LAC niosomes was further evaluated for permeation
Keywords: depth by confocal laser scanning microscopy (CLSM) and converted to gel formulation. Further, the
Lacidipine optimized LAC niosomes gel was evaluated for ex-vivo permeation study, skin irritation study, stability
Niosomes study and pharmacodynamics study. The optimized LAC niosomes formulation showed vesicle size,
Transdermal gel
entrapment efficiency and flux value of 676.98  10.92 nm, 82.77  4.34% and 38.43  2.43 mg/cm2/h
Confocal laser scanning microscopy
respectively, with spherical morphology. The comparative CLSM study showed that optimized LAC
Antihypertensive activity
niosomes formulation has shown maximum permeation (70.75 mm) as compare to LAC liposomes
formulation (58.26 mm). The optimized LAC niosomes gel showed skin permeation enhancement of 2.15
times as compare to control gel. Furthermore, in vivo antihypertensive activity showed significantly
higher (p < 0.001) reduction in blood pressure compared to oral suspension. Indeed, it was found that
niosomal vesicles represented to be an efficient nano vesicular carrier for transdermal delivery of
lacidipine.
© 2017 Published by Elsevier Masson SAS.

1. Introduction efficacy compared to other dihydropyridine calcium antagonists


[35]. It has a high degree of lipophilicity which deposits on the lipid
Cardiovascular disease is leading cause of mortality and moiety and continuously releases into the binding site during the
responsible for one-third of all deaths globally. The majority of clean up phase, so it has long duration of action [51]. It undergoes
cardiovascular disorders are caused not by single risk factor, but extensive first-pass hepatic metabolism which result to a low oral
rather a mixture of several factors like high levels of blood lipids, bioavailability (about 10%) [29].
obesity, physical inactivity, smoking, glucose intolerance/diabetes Several conventional and novel approaches have already
and age. High blood pressure (BP) certainly represents a explored for the formulation of lacidipine, including the using
amendable risk factor [10]. Lacidipine (LAC) is a dihydropyridine the nanoformulation [37], self nanoemulsifying system [32]
calcium-channel blocker developed for oral administration and lipotomes [33] and proniosomes [46] have reported limited
widely used in therapy since the early 1990s. It is used in the fruition. All these formulations have enhances the solubility and
treatment of hypertension and atherosclerosis and also possesses bioavailability of poorly water soluble drug lacidipine but also
an antioxidant effect [6,35]. In addition to calcium-channel reported for lower loading capacity and drug expulsion during
mediated vasodilation, it exhibits significantly higher antioxidant storage. But among these different approaches, the use of
niosomes as formulation act as a solubilizing matrix and offer
several advantages, modify the particle composition or surface to
adjust the drug release rate, acting as permeation enhancer,
* Corresponding author.
greater stability, or the affinity for the target site and longer shelf-
E-mail addresses: zzafarpharmacian@gmail.com (Ameeduzzafar),
sarimimam@gmail.com (S.S. Imam), alipharm786@gmail.com (A. Ali). life [24]. However, the is no specific literature report available on

http://dx.doi.org/10.1016/j.biopha.2017.06.043
0753-3322/© 2017 Published by Elsevier Masson SAS.
256 M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266

the lacidipine niosomal gel formulation using statistical design 2.2. Methods
approach for enhancing its antihypertensive activity on rat model.
Niosomes are vesicular nanocarriers and have gained much 2.2.1. Formulation and optimization of niosomes
attention as novel drug delivery systems in the last three decades LAC niosomes were formulated and optimized by the thin-film
due to their unique characteristics for transdermal application. hydration technique [48]. Precisely, cholesterol, Span 60, SPL 70
Niosomes are self-assembling non-ionic vesicle systems also called and LAC were taken in a clean and dry round bottom flask and the
as also called as non-ionic liposomes formed from nonionic lipid mixture was dissolved in chloroform: methanol mixture
surfactants in an aqueous environment. It has high potential to act (1:1 v/v). The organic solvent was removed by rotary evaporator
as carriers for poorly soluble drugs [7,41,25]. It is structurally above the lipid transition temperature. The traces of organic
similar to liposomes [43], but due to the better stability, they are solvents were removed under vacuum for the overnight period.
ideal alternative to liposomes in drug delivery systems [18,21]. The deposited thin lipid film was hydrated with phosphate buffer
They are biodegradable and biocompatible, relatively nontoxic and saline (PBS) with pH 7.4 by rotation at 150 rpm at room
capable of loading both hydrophilic and lipophilic drugs [9,36]. temperature. The resulted vesicles were hydrated for 2 h at room
Niosomes have lamellar structures composed of amphiphilic temperature to obtain large multilamellar vesicles (LMLV). To
molecules, known as surfactants, contain both hydrophobic (tails) prepare smaller vesicles, these vesicles were sonicated as
and hydrophilic (heads) groups and leading self-assembling summarized in Table 1 at 4  C at 40% output frequency (at
properties, aggregating into a variety of shapes like micelles or 40 W) (Titanium probe, Ultrasonicator, Model-UP100H, Hielscher
into a planar lamellar bilayer [42]. Hydrophilic drugs are entrapped Ultrasonics GmbH, Berlin, Germany). Similarly Rhodamine Red
inside of vesicular aqueous core or adsorbed on bilayer surfaces (RR) (0.03% w/v) loaded niosomes was prepared for confocal laser
while lipophilic substances are encapsulated by partitioning into microscopy study.
the lipophilic domain of the bilayers [48].
There are various nano drug delivery systems have been 2.2.2. Experimental design
reported as a promising approach to overcome the various The developed formulations were optimized using 4-factor 3-
drawbacks of conventional delivery system. These novel delivery level Box-Behnken statistical design. The rationale behind this
system have reported in permeability enhancement of the drug Box Behnken design based on the salient principles of Design of
across the stratum corneum of skin or cornea of eye and also experiments and quality by design (QbD) approach. The design was
enhances the bioavailability through different routes. The previous particularly selected because it needs less runs than a central
experimental studies carried out on diverse nano sized delivery composite design in cases of three or four independent variables. It
systems for various diseases like arthritis [19,26], hypertension provides understanding of the plausible interaction(s) among the
[3,29], schizophrenia [44,48], hepatoprotective activity [22,45], different levels of variables and helps in selecting “the best”
antifungal disease [47], Cancer [17,2] have vouched the utility of formulation with minimal expenditure of time, effort and
these delivery systems through different routes. In fact, niosomes developmental cost vis-a‘-vis the traditional one factor at a time
act as therapeutic reservoirs for drug delivery in a controlled (OFAT) approach [5]. The QbD methodology involves defining the
manner to improve bioavailability and enhanced therapeutic critical process parameters (CPPs) using screening and risk
efficacy over extended period of time [34]. assessment, optimization data analysis and optimum search
The current research work explored the feasibility of niosome through response surface methodology (RSM) to embark upon
as a carrier for the transdermal application of poorly soluble drug the design space and postulation of control strategy for continuous
Lacidipine. The developed formulations were optimized using improvement [38,4]. This property prevents a potential loss of data
four-factor three-level Box–Behnken statistical design and evalu- in those cases. The design matrix generated the nonlinear
ated for the vesicle size, shape, entrapment efficiency, in-vitro drug quadratic equation for the response as shown below;
release, transdermal flux, confocal laser scanning microscopy,
Y = b0 + b1A + b2B + b3C + b12AB + b13AC + b23BC + b11A2 + b22B2 +
histopathological examination and in-vivo antihypertensive effi-
b33C2 . . . . . . . . . .
cacy.
Where Y is the response related with each factor level combina-
2. Materials and methods tion; b0 is constant; b1, b2, b3 are linear coefficients, b12, b13, b23 are
interaction coefficients while b11, b22, b33 are quadratic coefficients
2.1. Materials generated from the observed experimental values of response from
experimental runs, while A, B and C are the coded intensity of
Lacidipine was received as gift sample from Unichem Pharma, independent variables. The terms A2, B2 and C2 (i = 1, 2 or 3)
Ahmadabad, India. Cholesterol, Span 60, Carbopol 934 and soya represent the interaction and quadratic terms, respectively [27].
phosphatidylcholine 70 (SPL 70) had been procured from The concentration range of independent variables along with their
Spectrochem, Mumbai, India and S.D. Fine Chemicals, Mumbai,
India respectively. All the other chemicals used for formulation Table 1
were of analytical grade and procured from S.D. Fine Chemical, Various independent and dependent variables used in LAC niosomes formulations
Mumbai, India. Albino Wistar rats (6–8 weeks/200–250 g) were by Box-Behnken design.
approved and provided by Institutional Animal Ethics Committee, Factor Level used, actual (coded)
and CPSCEA (Committee for the purpose of control and supervision
Independent variable Low ( 1) Medium (0) High (+1)
of experiments on animals) (1021/IAEC/2014), Govt of India,
Central Animal House of Hamdard University. Animal experiments A = span 60 (mM) 1 2 3
B = CHO (mM) 0.5 1.0 1.5
were carried out according to the Guidelines for the Care and Use of
C = hydration time (min) 45 52.5 60
Laboratory Animals that was approved by the IAEC of Jamia D = sonication time (min) 5 10 15
Hamdard. All the Albino Wistar rats were housed under controlled
environmental conditions (temperature, 25  2  C; humidity, Dependent variables Goal
55  15% RH, 12 h light/dark cycle) and were nourished with pellet Y1 = Vesicle size (nm) Minimize
Y2 = Entrapment efficiency (%) Maximize
diet (Lipton, Kolkata, India). Y3 = flux (mg/cm2/h) Maximize
M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266 257

low, medium and high levels were showed in Table 1. The selected process was repeated thrice to ensure that free drug was
independent variables for the experimental design were span 60 completely removed [48]. The% EE of niosomes formulations
concentration (A), cholesterol concentration (B), hydration time was calculated by the following equation.
(C) and sonication time (D) and their effect were observed on
%EE = (Total drug Drug in supernatant)/(Total drug)
vesicle size (Y1), entrapment efficiency (Y2) and flux (Y3) with five
centre point were shown in Table 2. The resulting experimental
values of the responses were compared with predicted value and 3.3. Ex-vivo skin permeation studies
linear regression plot between actual and predicted value of the
responses was plotted. The model was evaluated in terms of The permeation of LAC from niosomes and control was
statistically significant coefficient and R2 values and finally one measured through excised rat skin on diffusion cells. The study
optimum formulation was selected from point prediction method was performed using rat abdominal skin on a diffusion cell with an
and converted to gel for further study. effective diffusional area of 0.785 cm2 having 10 mL of receiver
capacity. The filled in receiver chamber. The receiver fluid
3. Characterization of niosomes formulation ethanolic: phosphate buffered saline (30:70% v/v, pH 7.4) was
stirred with a magnetic stirrer at a speed of 200 rpm, and the
3.1. Niosomes size and size distribution temperature was maintained 32  C  2  C in the whole study.
Ethanol was added in receiver sample to maintain sink conditions
The vesicles size and size distribution were determined by due to the extreme poor solubility in water [1]. 2 mL of the
dynamic light scattering (DLS) method, using a computerized niosomes formulation (equivalent to 4 mg) was placed into donor
inspection system (Zetasizer, HAS 3000; Malvern Instruments, compartment and covered with aluminum foil to provide occlusive
Malvern, United Kingdom). For vesicle size measurement, the conditions. Samples were withdrawn at regular intervals up to 48 h
vesicular suspension was further diluted with the phosphate buffer and analyzed for drug content by HPLC method [29]. Each
saline to avoid multiscattering events and the measurements were experiment was replicated three times and concentration of LAC
conducted in triplicate [12]. permeated through the skin was calculated.

3.2. Entrapment efficiency 3.4. Niosomes morphology

The entrapment efficiency (% EE) of LAC niosomes formulations Morphology of niosomes vesicle was visualized by using a
were determined by the centrifugal method. The formulations Morgagni 268D (Fei Electron Optics, Netherland) transmission
were centrifuged at 7000 rpm for 20 min and supernatant was electron microscope (TEM), with an accelerating voltage of 100 kV.
taken and diluted with PBS (pH 7.4). The drug concentration in the The sample for TEM observation was prepared by placing few
resulting solution was assayed by UV method at 240 nm. This drops of optimized niosomes dispersion that was previously

Table 2
Observed Box–Behnken Design LAC niosomes formulation runs with their actual and predicted experimental value of Y1 (particle size), Y2 (encapsulation efficiency), and Y3
(flux).

Runs Independent variables Dependent variables

A B C D (Y1) Mean  SD (Y2) Mean  SD (Y3) Mean  SD

Actual value Predicted value Actual value Predicted value Actual value Predicted value
LNF1 2.00 0.50 52.50 5.00 754.23  10.23 750.24 71.92  2.36 72.05 27.87  1.71 25.40
LNF 2 2.00 1.00 52.50 10.00 674.65  11.45 677.24 57.46  5.49 75.31 28.91  1.28 35.20
LNF 3 2.00 1.00 52.50 10.00 678.64  9.26 677.24 78.91  4.58 75.31 37.29  2.06 35.20
LNF 4 1.00 1.00 45.00 10.00 715.98  12.73 720.35 33.39  5.01 36.46 18.06  1.17 16.41
LNF 5 2.00 1.00 60.00 15.00 586.67  15.92 585.35 40.19  3.56 37.64 34.19  2.16 32.19
LNF 6 3.00 0.50 52.50 10.00 605.87  11.36 611.29 94.75  3.59 93.15 30.91  1.98 30.28
LNF 7 3.00 1.00 52.50 15.00 583.65  10.39 584.98 58.67  2.84 60.68 32.76  2.36 34.19
LNF 8 2.00 1.00 45.00 15.00 643.82  12.31 641.44 48.42  2.51 45.68 28.49  1.85 27.88
LNF 9 2.00 1.00 52.50 10.00 672.97  19.97 677.24 76.85  4.26 75.31 34.37  1.69 35.20
LNF10 2.00 0.50 45.00 10.00 613.98  15.84 608.59 66.28  3.49 67.05 14.56  2.54 15.87
LNF11 2.00 1.50 52.50 5.00 788.42  21.49 783.08 30.93  1.98 31.80 20.47  2.67 18.78
LNF12 3.00 1.00 52.50 5.00 697.92  13.63 703.15 58.98  2.27 60.66 21.73  1.78 24.13
LNF13 2.00 1.00 45.00 5.00 737.76  19.38 740.18 39.42  2.52 37.82 16.32  1.59 18.55
LNF14 1.00 1.50 52.50 10.00 760.38  16.29 756.07 36.68  3.65 34.12 17.84  1.79 18.70
LNF15 3.00 1.50 52.50 10.00 624.34  13.21 621.05 44.01  4.08 42.56 21.98  1.73 20.31
LNF16 2.00 0.50 60.00 10.00 648.45  11.62 643.31 66.85  4.13 69.31 35.65  2.53 34.20
LNF17 2.00 1.00 60.00 5.00 743.67  18.78 747.16 51.63  2.72 50.21 27.33  2.16 28.17
LNF18 2.00 1.00 52.50 10.00 682.98  14.23 677.24 80.54  3.24 75.31 36.98  2.35 35.20
LNF19 2.00 1.50 52.50 15.00 665.98  13.82 669.34 31.7  1.78 33.37 25.56  1.89 27.68
LNF20 2.00 1.50 45.00 10.00 712.57  16.79 717.24 30.93  2.11 30.82 21.26  1.74 22.83
LNF21 3.00 1.00 60.00 10.00 605.39  14.49 600.39 62.62  3.56 61.35 29.26  2.05 30.56
LNF22 1.00 1.00 52.50 5.00 812.45  20.27 810.65 39.99  2.89 40.33 21.65  1.87 20.33
LNF23 2.00 1.00 52.50 10.00 676.98  10.92 677.24 82.77  4.34 75.31 38.43  2.69 35.20
LNF24 1.00 1.00 52.50 15.00 673.98  9.25 668.28 34.92  2.59 35.59 25.91  2.52 23.62
LNF25 3.00 1.00 45.00 10.00 589.65  8.38 585.97 57.93  4.13 58.55 25.41  2.21 22.57
LNF26 1.00 1.00 60.00 10.00 653.76  10.43 656.81 36.82  2.38 38.01 19.85  1.95 22.35
LNF27 1.00 0.50 52.50 10.00 662.67  9.21 667.07 58.87  3.85 56.16 15.63  2.00 17.53
LNF28 2.00 0.50 52.50 15.00 598.71  10.47 603.42 64.82  4.27 65.76 28.52  2.45 29.86
LNF29 2.00 1.50 60.00 10.00 628.49  15.27 633.41 31.32  1.83 32.90 19.62  2.05 18.43

A = Cholesterol (mM); B = Span 60 (mM); C = Hydration time (min); D = Sonication time (min); Y1 = Particle size (nm); Y2 = Encapsulation efficiency (%); Y3 = Flux (mg/cm2/h).
258 M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266

Table 3 skin of the rat. The experiment was run in diffusion cell as
Summary of regression analysis for responses Y1 (particle Size in nm), Y2
described under ex-vivo skin permeation study for 8 h. Both the
(encapsulation efficiency in%), and Y3 (flux in mg/cm2/h) for fitting to different
models. skin sample was removed, washed with distilled water and tested
for probe penetration [8,48]. The excised nude rat skin was placed
Model R2 Adjusted R2 Predicted R2 SD %CV
on the microscopic slide with the SC side facing the cover glass. The
Response (Y1) CLSM (Olympus Fluo View; FV1000) was carried out with an argon
Linear 0.8151 0.7843 0.7111 28.73 –
laser beam with excitation at 488 nm and emission at 590 nm. The
2FI 0.8899 0.8288 0.6385 25.60 –
Quadratic 0.9956 0.9911 0.9768 5.82 0.87
skin sample was sliced into sections of 5–10 mm thickness through
the z–axis by CLSM with Fluo view software.
Response (Y2)
Linear 0.5854 0.5163 0.4897 12.78 – 3.6. Formulation of optimized LAC niosomes gel
2FI 0.6203 0.4093 0.3732 14.13 –
Quadratic 0.9484 0.8968 0.8878 5.90 10.92
The optimized LAC niosomes formulation (LNF23) was con-
Response (Y3) verted into gel to get epidermal retention for prolonged period of
Linear 0.3551 0.2476 0.1578 6.10 – time. The selection of optimized LAC niosomes formulation
2FI 0.4883 0.2041 0.0667 6.28 –
(LNF23) was done on the basis of formulation showed maximum
Quadratic 0.9046 0.8093 0.6270 3.07 11.78
transdermal flux. Briefly, Carbopol-R 940 as gelling agent (1% w/w)
was added into water and kept overnight for complete humecta-
diluted 50-fold with double-distilled water onto a 400-mesh tion of polymer chains. The optimized LAC niosomes formulation
carbon film coated copper grid. Before the carbon film dried on the (LNF23) equivalent to 2 mg of LAC (transdermal dose of LAC for
grid, it was negatively stained with 1% phosphotungstic acid for 48 h) was added to hydrate carbopol solution with continuous
10 s. The sample was dried in air before TEM observation [15]. stirring. Other ingredients like 15% w/v polyethylene glycol-400
(PEG-400) and triethanolamine (TEA) (0.5%w/v) were added to get
3.5. Confocal laser scanning microscopy (CLSM) study homogeneous dispersion of gel (optimized LAC niosomes gel).

Confocal laser scanning microscopy (CLSM) used to scan the 3.7. Characterization of optimized LAC niosomes gel
fluorescence signal of developed formulations at different skin
depths. The optimized LAC niosomes formulation and control 3.7.1. Viscosity
formulation (liposome) prepared with rhodamine red (RR-0.03%) The viscosity of optimized LAC niosomes gel was determined at
and were applied homogeneously and non-occlusive to the dorsal 25  2  C using Brook-field viscometer with spindle number C-50-

Fig. 1. Three-dimensional (A-B) and contour response surface plot (C-D) image showing influence of independent variables (A = Cholesterol; B = Span 60; C = Hydration time;
D = Sonication time) on particle size for LAC loaded niosomes.
M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266 259

1 (Brookfield Engineering Laboratories Inc., Middleboro, MA, USA), experiments were performed with conventional liposome. To
at 50 rpm and the determinations were carried out in triplicate. determine the extent of skin permeation enhancement, enhance-
ment ratio (ER) was calculated as follows:
3.7.2. pH
ER = steady state flux of formulation/steady state flux of control
The optimized LAC niosomes gel was diluted in double distilled
water and the pH was recorded using a pH meter (Mettler Toledo
MP 220, Greifensee, Switzerland) by bringing it in contact with the 3.7.5. Texture analysis of niosomal gel formulation
optimized niosomes gel and allowed it to stand for 1 min to Gel characteristics such as stickiness and firmness of LAC
equilibrate. niosomes gel were analyzed by using spreadability ring that was
fitted on the texture analyzer (Stable Micro Systems Ltd.,
3.7.3. Drug content Godalming, Surrey GU7 1YL, UK). It consists of two probes: an
For assay of LAC in niosomes gel, LAC was extracted from 10 g of upper probe and a lower probe. Niosomes gel was allowed to
optimized niosomes gel with 100 mL of methanol; it was suitably equilibrate for 30 min at room temperature before starting the test.
diluted and assayed by UV method at 240 nm. The analysis was The test sample was kept in the lower cone of the equipment and
performed triplicate for each batch and the content of LAC in each pressed to eliminate entrapped air and the excess was dragged off
sample was determined in terms of percentage of drug content. to make a flat test area. Before testing, the upper cone probe was
calibrated alongside the lower probe in such a way that the starting
3.7.4. Ex-vivo skin permeation study point can be kept at the same height. During testing, the
The skin permeation studies of optimized LAC niosomal gel was equipment was set to compression mode with a test speed of
performed on diffusion cell with an effective diffusional area of 3 mm/s. The upper cone probe was allowed to reach and penetrate
0.785 cm2and 10 mL of receiver chamber capacity using rat the sample up to a depth of 2 mm above the sample holder surface.
abdominal skin. After complete preparation and stabilization of Thus, the probe moved a distance of 23 mm from its starting point.
the skin 2gm of niosomal gel formulation was placed into donor
compartment and covered with aluminum foil to provide occlusive 3.7.6. In-vitro drug release and release kinetics
conditions. After specific time interval samples were withdrawn The release study was performed for the optimized niosomes
from receptor compartment at regular time intervals (1, 2, 4, 6, 12, gel formulation (LNF23) by the paddle method, using ethanol:
24, and 48 h), and analyzed for drug content by HPLC [29]. Similar phosphate buffer (30:70, pH 6.8) to simulate human skin pH. The

Fig. 2. Three-dimensional (A-B) and contour response surface plot (C-D) image showing influence of independent variables (A = Cholesterol; B = Span 60; C = Hydration time;
D = Sonication time) on encapsulation efficiency for LAC loaded niosomes.
260 M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266

calculated amount of niosomes gel formulation was placed in a classified on the basis of scoring. [PII <2 (non-irritant); PII = 2–5
cellulose membrane (MW = 12000 Da) and was placed at the (irritant); PII = 5–8 (highly irritant)]. Further skins were tested for
bottom of the USP dissolution tester (Veego, VDA-8DR, Mumbai, histopathological study by sacrificing Albino Wistar rat. The
India). The vessel contained 500 mL buffer solution and the speed treated area and untreated area were collected was stored in 10%
were adjusted to50 rpm [50]. About 5 mL were withdrawn from formalin solution in phosphate buffer solution. The samples were
the release medium at different time intervals for 24 h. and dehydrated using ethanol, embedded in paraffin and stained with
replaced by the equivalent volume of the medium. The amount of hematoxylin and eosin. The skin samples were then observed
drug released from the formulation was determined by HPLC. The under light microscope (Motic, Japan) and compared with the
mean cumulative amount of drug released was plotted against control sample and evaluated for any skin irritation effect.
time and data obtained from the release studies were kinetically
analyzed for the order of drug release from the formulation. The 3.7.8. In-vivo antihypertensive study
release evaluated to check the goodness of fit for zero-order model, The animals were trained for their stay in the restrainer because
first-order model, Higuchi's matrix model [13] and Korsmeyer– slight movement and aggression by the animal would have led to
peppas model [39]. The correlation coefficient (R2) for each model variation in BP reading. For this reason, a rat was inserted in the
was calculated. Model giving R2 nearer to 1 was selected as the best restrainer headlong until the whole body was easily accommodat-
fit model for the drug release. ed inside. The restrainer was locked from the open side and leaving
the tail outside the apparatus. The exercise was repeated several
3.7.7. Skin irritation and histopathological studies times for 7–10 days before the actual experiment until the animals
Skin irritation study was performed using Wistar rat to check learned to stay in the restrainer without aggression and was
for skin irritation potential [16]. After application of optimized LAC familiar with the experimental conditions. The animals (Wistar
niosomes gel formulation and aqueous formalin solution (positive rats) were divided into 4 groups (Group A to D) of 4 rats each.
control) [49,44], the application sites were examined for dermal Group A was taken as control and remaining three groups (Groups
reactions. The primary irritation score was calculated following B to D) hypertension was induced by subcutaneous injection of
study completion and compounds producing scores of 2 or less methylprednisolone acetate (MPA) (20 mg/kg/wk) for 2 weeks as
were considered no skin irritation. The primary irritancy index per reported method [23]). The BP of hypertensive rats from Group
(PII) was calculated for each treated group by adding the edema B to D was measured before the drug administration on the day of
and erythema scores of each group. The score was accordingly the actual experiment. Group B served as toxic control and received

Fig. 3. Three-dimensional (A-B) and contour response surface plot (C-D) image showing influence of independent variables (A = Cholesterol; B = Span 60; C = Hydration time;
D = Sonication time) on flux for LAC loaded niosomes.
M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266 261

Table 4
In vitro release kinetics data to different mathematical models for LAC loaded optimized niosomes and optimized niosomes gel formulations.

Optimized niosomes (LAC23) Optimized niosomal gel (LAC23 Gel)


2
Model Equation R Model Equation R2
Zero-order mo m = kt 0.8908 Zero-order mo m = kt 0.9391
First-order ln m = kt 0.9606 First-order ln m = kt 0.9773
Higuchi matrix mo m = kt1/2 0.9744 Higuchi matrix mo m = kt1/2 0.9962
Korsmeyer- peppas log (mo m) = log k + n logt 0.9774 Korsmeyer- peppas log (mo m) = log k + n logt 0.9826
Best fit model Korsmeyer- peppas Best fit model Higuchi model

no more treatment, Group C received an oral suspension of LAC 4. Results and discussion
(1 mg/kg) with the help of feeding needle and group D received
optimized LAC niosomes gel formulation, respectively [11,40]. The 4.1. Formulation and optimization of LAC niosomes
formulation was applied on the previously shaven abdominal area
of rat skin. The rat was then placed in the restrainer and the BP A 4-factor, 3-level Box–Behnken statistical design was used to
from the tail was recorded at predetermined time intervals up to preparation niosomes (Table 2) using Design Expert1 (Version
48 h (Table 3). 8.0.0.6, Stat-Ease Inc., Minneapolis, MN). All individual and
interactive effects of independent variables were investigated
3.7.9. Stability study of LAC niosomes gel formulation and all the responses of these runs fitted to first order, second order
The optimized LAC niosomes gel formulation was subjected to and quadratic models and found that best fit model was quadratic
stability studies to evaluate any physical or chemical changes in (p < 0.0001). The Summary of results of regression analysis for
storage. Gel formulation was kept at 25  2  C/60  5% RH and responses Y1, Y2 and Y3 for fitting to quadratic model and the
40  2  C/75  5% RH for 1 month in borosil glass container ICH polynomial equation of each response and each model were shown
Q1A (R2), [14]. The samples were analyzed for various parameters in Table 3. Three-dimensional plots showed the interaction effects
at 0, and 1 month after storage like Clarity, pH, drug content and of the independent variables on the responses as well as their
spreadability. usefulness in studying the effects of two factors on one response at
a time (Figs. 1–3).

Fig. 4. Linear correlation plots between actual and predicted values and the corresponding residual plots for all responses.
262 M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266

4.1.1. Fitting of data to the model The Model F-value of 224.63 implies the model is significant.
The results of regression analysis of different responses are There is only a 0.01% chance that a “Model F-Value” this large could
given in Table 4. Larger values of the standard error for coefficients occur due to noise. Values of “Prob> F” less than 0.05 indicated that
shows that the quadratic nature of the relationship. From Table 2, it the model terms are significant. In this case, A, B, C, D, AB, AC, BC,
is evident that the one independent variables viz. the concentra- BD, CD, A2, C2, D2 are significant model terms. Values greater than
tion of the Span 60 have positive effects on the response Y2 0.1 indicates the models are not significant. The “Lack of Fit F-
(entrapment efficiency) and response Y3 (flux), whereas the value” of 2.77 implies the “Lack of Fit” is not significant which is
response Y1 (vesicle size) has an inverse relationship with Span relative to the pure error. There is only 16.91% chance that this large
60. The concentration of cholesterol has the positive effect on “Lack of Fit F-value” could occur due to noise. There existed a direct
response Y1 (vesicle size) whereas the concentration of cholesterol relationship between the Span 60 concentration and cholesterol on
has inverse effect on response Y2 (entrapment efficiency) and on the vesicle size, entrapment efficiency of the vesicles and
response Y3 (flux). Hydration time and sonication time have an transdermal flux of vesicles loaded with LAC. This model can be
inverse effect on response Y1 (vesicle size) whereas both have a used to navigate the design space and result of this calculated
positive effect on response Y3 (flux). model for vesicle size represented by 3-dimentional plots and
contour plot as shown in Fig. 1 (A-D). The plot showed the effect of
4.1.2. Effect of independent variables on size two formulation factors on particle size at one time.
Small vesicular size is most important criteria for the effective
transdermal drug delivery of niosomes. The size of the vesicles was 4.1.3. Effect of independent variables on EE
found to vary between 583.65 to 812.45 nm (Table 2). Initially, Entrapment efficiency (%EE) is the percentage fraction of the
average vesicle size increased with increase in the concentration of total drug (LAC) entrapped into the vesicles. The maximum and
Span 60 from 1 to 2 mM. However, an additional increase in the minimum entrapment efficiency obtained were 94.75% for LNF6
concentration of Span 60 from 2 to 3 mM leads to decrease in the and 30.93% for LNF20 respectively (Table 2). It is observed from the
average vesicle size. This is due to the development of a micellar experimental design that entrapment efficiency has a direct
structure instead of the vesicles, which are comparatively smaller positive relationship with the concentration of Span 60 as revealed
in size. This relationship is presented by the following equation. by the following equation.
Y1 (Particle Size) = 677.244 47.7A + 24.6892B 12.2775C Y2 (Entrapment Efficiency) = 75.306 + 11.3575A 18.16
65.1367 D 19.8 AB + 19.49 AC + 6.05 AD 29.6375 BC + 8.27 B + 1.08833C 1.17917 D 7.1375 AB + 0.315 AC + 1.19 AD 0.045
BD 15.765 CD 11.5666 A2 1.81033 B2 24.7978 C2 + 26.0859 BC + 1.9675 BD 5.11 CD 10.1176 A2 8.6888 B2 16.5963
D2 C2 15.8726 D2

Fig. 5. Optimized LAC niosomes (a) Particle size graph (b) Transmission electron micrograph (80,000) (scale bar indicates in nm).
M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266 263

The Model F-value of 18.38 implies the model is significant. D2 are significant model terms. The values greater than 0.1
There is only a 0.01% chance that a “Model F-Value” this large could indicated that the model terms are not significant. The “Lack of Fit
occur due to noise. Values of “Prob> F” less than 0.05 indicated that F-value” of 0.51 implies that this small “Lack of Fit F-value” is not
the model terms are significant. In this case, A, B, AB, A2, B2, C2, D2 significant which is relative to the pure error. There is 82.42%
are significant model terms. Values greater than 0.1 indicated that chance that this large “Lack of Fit F-value” could occur due to noise.
the model terms are not significant. The “Lack of Fit F-value” of 0.07 Effects of Independent variables on flux are presented by three-
implies the “Lack of Fit” is not significant which is relative to the dimensional graph and contour plot Fig. 3 (A-D).
pure error. There is 99.97% chance that this large “Lack of Fit F-
value” could occur due to noise. This model can be used to navigate 4.2. Optimization
the design space and result of this calculated model for entrapment
efficiency represented by 3-dimentional plots and contour plot as The optimized LAC loaded niosomal formulation was selected
shown in Fig. 2 (A-D). As the concentration of Span 60 increases, % based on the criteria of attaining the maximum value of
EE also increases. It was found that upon increasing hydration time transdermal flux and entrapment efficiency whereas minimizing
the% EE significantly increased from 36.82% (LNF26, with least the vesicle size by applying point prediction method of the Design
concentration cholesterol and Span 60) to 71.92% LNF1, medium Expert1 software [30]. Upon ‘trading of’ various response variables
ratio cholesterol: Span 60 (2:0.5). According to [28] the entrap- and comprehensive evaluation of feasibility search and exhaustive
ment of drug occurred in both, bilayer and aqueous compartment grid search, the formulation composition with Span 60 (2 mM),
of the vesicles. When the lipid compartment and aqueous phase cholesterol (1 mM), hydration time (52.5 min) and sonication time
becomes saturated with the drug, the vesicles provided limited (10 min) was found to fulfil requisites of an optimum formulation
entrapment capacity. The entrapment of drug occurs in both the i.e. LNF 23. The optimized formulation has the vesicle size of
bilayers and the aqueous compartment of the vesicles [20]. When 676.98  10.92 nm with entrapment efficiency 82.77 4.34% and
the lipid compartment and aqueous phase became saturated with transdermal flux across rat skin of 38.43 2.69 mg/cm2/h,
the drug, the vesicles provided limited entrapment capacity [31]. respectively. Fig. 4 showed the quantitatively linear relationship
Hence, niosome could entrap LAC only to an optimum extent, after between resultant experimental values of the responses with that
which any further increase in hydration time would lead to leakage of the predicted value of all dependents variables. Optimized LAC
of LAC from vesicles. niosomes formulation (LNF 23) was converted into gel formulation
and applied transdermally for in vivo antihypertensive studies.
4.1.4. Effect of independent variables on flux
There is no significant effect of hydration time on transdermal 4.3. Characterization and evaluation of niosomes
flux. The maximum and minimum transdermal flux were found to
be 38.43 (LNF 23) and 14.56 (LNF10) as shown in Table 2. It was 4.3.1. Vesicles size and size distribution
observed from the equation below that the transdermal flux of LNF The least mean vesicles size was observed for LAC loaded
has a direct relationship with the concentration of cholesterol niosomes formulations LNF7 (583.65 nm) while maximum vesicles
content as revealed by the following equation. size was obtained as 812.45 nm for LNF22 shown in Table 2. The
Y3 (Flux) = 35.196 + 3.5925 A 2.2008 B + 3.4833C + 3.3383 D size of optimized formulation was found 683.8 nm and PDI was
2.785 AB + 0.515 AC + 1.692 AD 5.682 BC + 1.11 BD 1.327 found 0.329 as shown in Fig. 5a.
CD 6.675 A2 6.8155 B2 5.5492 C2 2.9492 D2
The Model F-value of 9.49 implies that the model is significant. 4.3.2. Vesicles shape
There is only a 0.01% chance that this large “Model F-Value” could The transmission electron micrograph (TEM) of the optimized
occur due to noise. Values of “Prob> F” less than 0.05 indicated that niosomes formulation (LNF23) is shown in Fig. 5b. TEM micro-
the model terms are significant. In this case, A, B, C, D, BC, A2, B2, C2, graph revealed the formation of well identified spherical and

Fig. 6. CLS micrographs of rat skin cross section images after application with rhodamine red loaded (0.3%) (a) optimized LAC niosomes gel formulation (b) LAC liposomal gel
formulation (control) (Scale bar indicates in mm). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
264 M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266

Fig. 7. Haematoxylin and eosin stained cross section photomicrograph of rat skin (a) untreated (control), (b) formalin treated (c) optimized LAC niosomes gel treated.

Table 5
Comparative pharmacodynamic assessment on mean BP of various formulation of LAC in MPA induced hypertensive rat.

Group Treatment Systolic BP (mm Hg) % reduction in BP

0h 1h 2h 4h 6h 12 h 24 h 48 h
A N saline 115.4  5.4 116.3  9.2 115.2  7.1 114.6  4.6 118.2  6.3 115.7  5.1 117.3  4.9 118.2  5.6
B HC control 182.2  7.9 185.8  8.1 178.2  6.1 186.5  6.9 180.1  7.1 181.8  7.3 189.5  8.1 181.5  5.9
C OLS suspension 180  7.5 142.3  8.1 122.5  6.7 136.2  5.4 145.5  6.9 154.1  7.3 159.3  5.4 161.9  6.8 10.79
D OLNniosomal gel 181.2  7.5 161.6  5.9 143.5  8.1 134.6  5.1 126.2  4.9 122.3  4.1 118.7  3.9 141.5  6.4 22.04

NS:- normal saline; HC: Hypertensive control; OLS: Oral LAC Suspension; OLN: optimized LAC niosomal.

discrete vesicles with sharp boundaries after hydration of 4.3.6. Ex vivo permeation study
niosomes. It shows the outline and core of the well identified The ex-vivo permeation profile of niosomes yielded significantly
spherical vesicles, and displaying the retention of sealed vesicular higher (p < 0.0001) flux value i.e. 38.43 mg/cm2/h (optimized LNF
structure. The surface morphology of niosomes formulation 23 gel) with enhancement ratio of 33.92 through rat skin. The
presents increased vesicle wall rigidity with increasing cholesterol reason for this better performance of niosomes formulations is the
content. deformability and the ability to retain vesicle integrity, while the
aggregates undergo a dramatic change in shape in comparison
4.3.3. Confocal laser scanning microscopy (CLSM) study with liposomes and all these characteristics allow the niosomes to
The comparative CLSM study between rhodamine loaded LAC pass the skin pores which are much smaller than the diameter of
niosomes formulation and liposome formulation (control) was niosomes [40]). Effects of independent variables on flux are
performed to check the penetration depth. The result revealed that presented by three-dimensional graph and contour plot as shown
rhodamine red loaded LAC niosomes formulation was uniformly in (Fig. 3).
distributed to a greater extent throughout the subcutaneous, viable
epidermis, and dermis with high fluorescence intensity [50]. The 4.3.7. In-vitro drug release and release kinetics
formulation exhibited significantly higher depth (70.75 mm) of The obtained in vitro release data for both optimized LAC
penetration compared to control formulation (38.25 mm) after niosomes and LAC niosomes gel data was fitted to various release
transdermal application as depicted in Fig. 6 (A-B). The promi- kinetic models. The correlation coefficients (R2) values for different
nently efficient delivery of LAC by niosomal carriers suggested models for both formulations are presented in Table 4. The model
their enhanced penetration and consequent fusion with the showing highest value of R2 was considered as best model for
membrane lipids in the depths of the skin, corroborating the release mechanism. It was found that the best fit model for
results of many researchers [8]. optimized LAC niosomes (LAC23) was Korsmeyer-peppas model
and for LAC niosomes gel was Higuchi matrix model, respectively.
4.3.4. Characterization of optimized LAC niosomal gel The highest R2 obtained for optimized LAC niosomes gel
The drug content, viscosity, pH and spreadability of optimized formulation (LAC23) and optimized LAC niosomes gel formulation
niosomal gel were shown 98.60%, 38.00 PaS, 6.7 and satisfactory were 0.9744 and 0.9962 respectively.
spreadability, respectively.
4.3.8. Histopathological study
4.3.5. Texture of niosomal gel formulation Sections of untreated rat skin (control) revealed well defined
The various parameter of texture analysis like of were like epidermal and dermal layers. The keratin layer was also well
consistency, stickiness, firmness and work of adhesion were formed and was presenting just adjacent to the top most layer of
evaluated of optimized LAC niosomal gel. The consistency, the epidermis. Dermis was devoid of any inflammatory cells as
stickiness, firmness and adhesion of optimized LAC-niosomal gel clearly seen in Fig. 7(A-C). There were no significant changes
was found to be 0.029  0.003, 0.032  0.002, 0.048  0.003 and observed in rat skin specimens treated with formulation in
0.018  0.003 respectively. comparison with the untreated SC suggest absence of any skin
M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266 265

irritation. The formalin treated skin showed marked damage to the also grateful to AICTE, New Delhi, India, for providing senior
skin. The image shows marked damage to skin and extraction of research fellowship.
lipid has been seen in the formalin treated skin.
References
4.3.9. In vivo studies
The pharmacodynamic data of different treatments were [1] A. Ahad, M. Aqil, K. Kohli, Y. Sultana, M. Mujeeb, A. Ali, Formulation and
optimization of nanotransfersomes using experimental design technique for
compared for statistical significance by Dunnet test, which showed accentuated transdermal delivery of valsartan, Nanomedicine 8 (2012) 237–
significant difference (P < 0.001) in BP values of normal control 249.
(Group A) (Table 5). The oral administration of LAC suspension [2] A. Khan, S.S. Imam, M. Aqil, A. Ahad, Y. Sultana, A. Ali, K. Khan, Brain targeting
of temozolomide via the intranasal route using lipid-based nanoparticles:
significantly (p < 0.05) controlled the hypertension within 2 h, brain pharmacokinetic and scintigraphic analyses, Mol. Pharm. 13 (2016)
with the maximum antihypertensive effect (122 mm Hg), beyond 3773–3782.
2 h mean systolic blood pressure remained above the normal value [3] A. Mishra, S.S. Imam, M. Aqil, A. Ahad, A. Ali, Carvedilol Nano lipid carrier:
formulation, characterization and in-vivo evaluation, Drug Deliv. 23 (4) (2016)
and ultimately reached to 161 mm Hg at 48 h. Oral suspension of
1486–1494.
LAC acted rapidly and showed utmost changes in rat BP, but its [4] B. Singh, R. Kapil, M. Nandi, N. Ahuja, Developing oral drug delivery systems
effect dipped rapidly and BP increased to above normal value. The using formulation by design: vital precepts, retrospect and prospects, Expert
Opin. Drug Deliv. 8 (2011) 1341–1360.
administration of LAC oral suspension resulted in a gradual
[5] B. Singh, R. Kumar, N. Ahuja, Optimizing drug delivery systems using
decrease of BP, with the maximum effect of 122.5 mm Hg, observed systematic “design of experiments. ” Part I: Fundamental aspects, Crit. Rev.
at 6 h on treatment the experimental hypertensive rats (p > 0.05). Ther. Drug Carrier. Syst. 22 (2005) 27–105.
The result revealed that the systolic BP of hypertensive rats was [6] C.R. Lee, H.M. Bryson, Lacidipine A review of its pharmacodynamic and
pharmacokinetic properties and therapeutic potential in the treatment of
controlled up to 48 h; however at, 48 h time point BP, was hypertension, Drugs 48 (1994) 274–296.
comparable to that of the normotensive rats (p > 0.05). The [7] C.T. Lo, A. Jahn, L.E. Locascio, W.N. Vreeland, Controlled self-assembly of
decrease in systolic blood pressure by LAC niosomes gel was monodisperse niosomes by microfluidic hydrodynamic focusing, Langmuir 26
(2010) 8559–8566.
gradual, not sharp as with oral suspension, because of controlled [8] E. Touitou, N. Dayan, L. Bergelson, B. Godin, M. Eliaz, Ethosomes-novel
release of LNF. The systems were effective and could control the BP vesicular carriers for enhanced delivery: characterization and skin penetration
up to 48 h. The above results are in conformity with ex vivo release properties, J. Control. Rel. 65 (3) (2000) 403–418.
[9] E.R. Bendas, H. Abdullah, M.H.M. El-Komy, M.A.A. Kassem,
study data. The above result suggested that the developed LAC Hydroxychloroquine niosomes: a new trend in topical management of oral
niosomes gel formulation hold promise for the management of lichen planus, Int. J. Pharm. 458 (2013) 287–295.
hypertension that needs to be validated by in vivo study and [10] G. Mancia, G. Grassi, S.E. Kjeldsen, Nadcisnienietetnicze podrecznik
European Society of Hypertension, VIA, MEDICA, Gdansk, 2009.
clinical trial.
[11] G.E. Paget, J.M. Barnes, in: D.R. Laurence, A.L. Bacharach (Eds.), Evaluation of
Drug Activities: Pharmacometrices, vol.1, Academic press, New York and
4.3.10. Stability studies London, 1964.
[12] G.M.M. El-Maghraby, A.C. Williams, B.W. Barry, Skin hydration and possible
The influence of elevated temperature and humidity conditions
shunt route penetration in controlled oestradiol delivery from
on the physical stability of LAC niosomes gel formulation was ultradeformable and standard liposomes, J. Pharm. Pharmacol. 53 (2001)
performed in adherence to the ICH guidelines. After period of 0, 1311–1322.
and 1 month clarity, pH, homogeneity and drug content were [13] T. Higuchi, Mechanism of sustained- action medication. Theoretical analysis of
rate of release of solid drugs dispersed in solid matrices, J. Pharm. Sci. 52
determined in the formulation. In both storage conditions the (1963) 1145–1149.
formulation remained physically stable. No significantly (p > 0.001) [14] ICH Topic Q 1 A (R2), Stability Testing of New Drug Substances and Products
changes were observed. The shelf life of the formulation was found CPMP/ICH/2736/99, (2003) (August).
[15] J. Guo, Q. Ping, G. Sun, C. Jiao, Lecithin vesicular carriers for transdermal
to be approx 1.8 h with degradation rate constant delivery of cyclosposrine, Int. J. Pharm. 194 (2000) 201–207.
[16] J.H. Draize, G. Woodard, H.D. Calvery, Methods for the study of irritation and
5. Conclusion toxicity of substances applied topically to the skin and mucous membranes, J.
Pharmacol. Exp. Ther. 82 (1944) 377–390.
[17] K. Wang, Q. Huang, F. Qiu, M. Sui, Nonviral delivery systems for the application
The present study conclusively demonstrates the use of Box- in p53 cancer gene therapy, Curr. Med. Chem. 22 (35) (2015) 41184136.
Behnken design in formulation and optimization of LAC niosomes [18] K.M. Kazi, A.S. Mandal, N. Biswas, A. Guha, S. Chatterjee, M. Behera, K. Kuotsu,
Niosome: a future of targeted drug delivery systems, J. Adv. Pharm. Technol.
gel formulations to avoid its systemic toxicity. This study indicated Res. 1 (2010) 374–380.
that niosomes can be optimized to achieve desired properties [19] L. Abidin, M. Mujeeb, S.S. Imam, M. Aqil, D. Khurana, Enhanced transdermal
using span 60 concentrations, cholesterol concentrations, sonica- delivery of Luteolin via non-ionic surfactant based vesicle: quality evaluation
and antiarthritic assessment, Drug Deliv. 23 (3) (2016) 1069–1074.
tion time, and hydration time. The optimized niosomes formula-
[20] L.B. Lopes, M.V. Scarpa, G.V.J. Silva, D.C. Rodrigues, C.V. Santilli, A.G. Oliveira,
tion demonstrated spherical morphology, enhanced entrapment Studies on the encapsulation of Diclofenac in small unilamellar liposomes of
efficiency, and reasonable polydispersity index and particle size. soya phosphatidylcholine, Colloids. Surf. B: Biointerfaces 39 (2004) 151–158.
Confocal laser scanning microscopy further confirmed the poten- [21] L.D. Marzio, C. Marianecci, M. Petrone, F. Rinaldi, M. Carafa, Novel pH-sensitive
non-ionic surfactant vesicles: comparison between Tween 21 and Tween 20,
tial of the developed formulation by showing the high fluorescent Colloids Surf. B 82 (2011) 18–24.
intensity in comparison to conventional liposomes. The optimized [22] M. Akhtar, S.S. Imam, M. Afroz, M. Mujeeb, A.K. Najmi, M. Aqil, Neuroprotective
LAC niosomal gel formulation was found to decrease the BP study of Nigella sativa loaded oral provesicular lipid formulation: in-vitro and
ex-vivo study, Drug Deliv. 21 (6) (2014) 487–494.
significantly in experimental hypertensive rats, which was [23] M. Aqil, A. Ali, Y. Sultana, K. Dubey, A.K. Najmi, K.K. Pillai, In vivo
maintained for 48 h. The developed niosomes system was found characterization of monolithic matrix type transdermal drug delivery
to be a good carrier for LAC by transdermal delivery. systems of pinacidil monohydrate: a technical note, AAPS PharmSciTech 7 (1)
(2006) E6.
[24] M. Bragagni, A. Scozzafava, A. Mastrolorenzo, C.T. Supuran, Paola Mura
Conflict of interest Development and ex vivo evaluation of 5-aminolevulinic acid-loaded
niosomal formulations for topical photodynamic therapy, Int. J. Pharm. 494
(2015) 258–263.
Authors have no conflict of interest [25] M. Bragagni, N. Mennini, S. Furlanetto, S. Orlandini, C. Ghelardini, P. Mura,
Development and characterization of functionalized niosomes for brain
Acknowledgments targeting of dynorphin-B, Eur. J. Pharm. Biopharm. 87 (2014) 73–79.
[26] M. Jamal, S.S. Imam, M. Aqil, M. Amir, S.R. Mir, M. Mujeeb, Transdermal
potential and anti-arthritic efficacy of ursolic acid from niosomal gel systems,
The authors are thankful to Unichem Pharma Pharmaceuticals Int. Immunopharmacol. 29 (2015) 361–369.
Ltd., India for providing gift sample of carteolol. The authors are
266 M. Qumbar et al. / Biomedicine & Pharmacotherapy 93 (2017) 255–266

[27] M. Khajeh, Application of Box-Behnken design in the optimization of a [40] S. Jain, B.H. Chaudhari, N.K. Swarnakar, Preparation and characterization of
magnetic nanoparticle procedure for zinc determination in analytical samples niosomal gel for iontophoresis mediated transdermal delivery of
by inductively coupled plasma optical emission spectrometry, J. Hazard Mater. isosorbidedinitrate, Drug Deliv. Transl. Res. 1 (2011) 309–321.
172 (1) (2009) 385–389. [41] S. Kumar, D. Bharjava, A. Thakkar, S. Arora, Drug carrier systems for solubility
[28] M. Mokhtar, O.A. Sammour, M.A. Hammad, N.A. Megrab, Effect of enhancement of BCS class II drugs: a critical review, Crit. Rev. Ther. Drug
someformulation parameters on flurbiprofen encapsulation and release Carrier Syst. 30 (2013) 217–256.
rates of niosomes prepared from proniosomes, Int. J. Pharm. 361 (2008) 104– [42] S. Moghassemi, A. Hadjizadeh, Nano-niosomes as nanoscale drug delivery
111. systems:an illustrated review, J. Control. Release 185 (2014) 22–36.
[29] M. Qumbar, Ameeduzzafar, J. Ali, S.S. Imam, Mohd Fazil, A. Ali, DOE-Based [43] S. Moghassemi, E. Parnian, A. Hakamivala, M. Darzianiazizi, M.M. Vardanjani,
stability indicating RP-HPLC method for determination of lacidipine in S. Kashanian, B. Larijani, K. Omidfar, Uptake and transport of insulin across
niosomal gel in rat: pharmacokinetic determination, Pharm. Anal. Acta 5 intestinal membrane model using trimethyl chitosan coated insulin niosomes,
(2014) 8. Mater. Sci. Eng. C 46 (2015) 333–340.
[30] A. Ahad, M. Aqil, K. Kohli, Y. Sultana, M. Mujeeb, Enhanced transdermal [44] S.S. Imam, M. Aqil, A. Ahad, M. Akhtar, Y. Sultana, A. Ali, Formulation by design
delivery of an anti-hypertensive agent via nanoethosomes: Statistical based risperidone nano soft lipid vesicle as a new strategy for enhanced
optimization, characterization and pharmacokinetic assessment, Int. J. of transdermal drug delivery: in-vitro characterization, and in-vivo appraisal,
Pharm. 443 (2013) 26–38. Mater. Sci. Eng. C 75 (2017) 1198–1205.
[31] M.Y. Ning, Y.Z. Guo, H.Z. Pan, H.M. Yu, Z.W. Gu, Preparation and evaluation of [45] S. Sayeed, S.S. Imam, A.K. Najmi, M. Aqil, M. Akhtar, Nonionic surfactant based
proliposomes containing clotrimazole, Chem. Pharm. Bull. (Tokyo) 53 (2005) thymoquinone loaded nanoproniosomal formulation: in vitro
620–624. physicochemical evaluation and in vivo hepatoprotective efficacy, Drug Dev.
[32] N. Subramanian, S.P. Sharavanan, P. Chandrasekar, A. Balakumar, S.P. Moulik, Ind. Pharm. (2017), doi:http://dx.doi.org/10.1080/03639045.2017.1318903.
Lacidipine self nanoemulsifying drug delivery system for the enhancement of [46] S.M. Soliman, N.S. Abdelmalak, O.N. ElGazayerly, N. Abdelaziz, Novel nonionic
oral bioavailability, Arch. Pharm. Res. 39 (4) (2016) 48191. surfactant proniosomes for transdermal delivery of lacidipine: optimization
[33] N.A. El Kasabgy, I. Elsayed, A.H. Elshafeey, Design of lipotomes as a novel dual using 2(3) factorial design and in vivo evaluation in rabbits, Drug Deliv. 23 (5)
functioning nanocarrier for bioavailability enhancement of lacidipine: in-vitro (2016) 160822.
and in-vivo characterization, Int. J. Pharm. 472 (1–2) (2014) 369–379. [47] S.R.M. Moghddam, A. Ahad, M. Aqil, S.S. Imam, Y. Sultana, Formulation and
[34] P.K. Lakshmi, S. Bhaskaran, Phase II study of topical niosomal urea gel an optimization of niosomes for topical diacerein delivery using 3-factor:3-level
adjuvant in the treatment of psoriasis, Int. J. Pharm. Sci. Rev. Res. 7 (2011) 1–7. Box-Behnken design for the management of psoriasis, Mater. Sci. Eng. C 69
[35] P.L. McCormack, A.J. Wagstaff, Lacidipine A review of its use in the (2016) 789–797.
management of hypertension, Drugs 63 (2003) 2327–2356. [48] S.S. Imam, M. Aqil, M. Akhtar, Y. Sultana, A. Ali, Formulation by design-based
[36] Q. Li, Z. Li, W. Zeng, S. Ge, H. Lu, C. Wu, L. Ge, D. Liang, Y. Xu, Proniosome derived proniosome for accentuated transdermal delivery of risperidone: in vitro
niosomes for tacrolimus topical ocular delivery: in vitro cornea permeation, characterization and in vivo pharmacokinetic study, Drug Deliv. 22 (8) (2015)
ocular irritation, and in vivo anti-allograft rejection, Eur. J. Pharm. Sci. 62 1059–1070.
(2014) 115–123. [49] U. Ubaidulla, M.V.S. Reddy, K. Ruckmani, F.J. Ahmad, R.K. Khar, Transdermal
[37] C. Shirodkar, G.H. Misra, P. Chethan, Z. Shetty, S. Attari, S. Mutalik, Subacute therapeutic system of carvedilol: effect of hydrophilic and hydrophobic matrix
toxicity profile of lacidipine nanoformulation in wistar rats, Sci. World J. (2015) on in-vitro and in-vivo characteristics, AAPS Pharm. Sci. Tech. 8 (2009) 1065–
(12 pages). 1070.
[38] R.A. Lionberger, S.L. Lee, L. Lee, et al., Quality by design: concepts for ANDAs, [50] B. Junyaprasert, P. Singhsa, A. Jintapattanakit, Influence of chemical
AAPS J. 10 (2008) 268–276. penetration enhancers on skin permeability of ellagic acid-loaded
[39] R.W. Korsmeyer, R. Gurny, E. Doelker, P. Buri, N.A. Peppas, Mechanisms of niosomes, Asian J. Pharm. Sci. 8 (2013) 110–117.
solute release from porous hydrophilic polymers, Int. J. Pharm. 15 (1983) 25– [51] Y.L. Geng, J. Zhao Guo, B.P. Ma, Development of a supercritical fluid
35. chromatography-tandem mass spectrometry method for the determination
of lacidipine in beagle dog plasma and its application to a bioavailability study,
J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 15 (945–946) (2014) 21–26.

You might also like