You are on page 1of 11

http://informahealthcare.

com/lpr
ISSN: 0898-2104 (print), 1532-2394 (electronic)

J Liposome Res, Early Online: 1–11


! 2014 Informa Healthcare USA, Inc. DOI: 10.3109/08982104.2014.974058

RESEARCH ARTICLE

Preparation and evaluation of a buflomedil hydrochloride niosomal


patch for transdermal delivery
Nida Akhtar1, Seema Arkvanshi1, Shiv Sankar Bhattacharya1, Anurag Verma1, and Kamla Pathak2
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14

1
School of Pharmaceutical Sciences, IFTM University, Lodhipur-Rajput, Moradabad, Uttar Pradesh, India, and 2Department of Pharmaceutics,
Rajiv Academy for Pharmacy, P.O. Chhattikara, Mathura, Uttar Pradesh, India

Abstract Keywords
Context: Niosomes are the non-ionic surfactant vesicles obtained on hydration of synthetic Cholesterol, drug, flux, permeability, systemic
non-ionic surfactants. These are the promising vehicles for effective transdermal drug delivery. circulation
Objective: The present research work was aimed to develop niosomal-based transdermal
buflomedil hydrochloride patch containing a stable formulation with improved drug History
permeation.
Materials and methods: Niosomes were prepared by solvent evaporation method using 32 Received 27 May 2014
factorial design. All the formulations were evaluated for vesicle size, zeta potential and percent Revised 30 August 2014
entrapment efficiency. Optimized niosomal and liposomal formulation were loaded into a Accepted 4 October 2014
patch system. All the patches were then characterized for drug–excipient interaction study, Published online 30 October 2014
scanning electron microscopy, pharmacotechnical properties and in vitro permeation studies.
For personal use only.

Result: F9 formulation having optimum vesicle size (10.09 ± 1.2 mm), highest zeta potential
(85.4 ± 0.56 mV) and maximum percent entrapment efficiency (97.09 ± 0.11%) was selected as
optimized formulation. In case of liposomes, formulation F12 was selected. Patches loaded
with niosomes showed 95.12 ± 1.19% cumulative amount of drug permeated as compared
to liposomal vesicle-loaded patches which showed 82.21 ± 1.24% and control patches
70.10 ± 1.33%. Discussion: Flux, permeation rate and permeability coefficient were found to
be higher in case of niosomal patches as compared to liposomal patches and control patches.
Surfactant present in niosomes act as a penetration enhancer which contribute in the
permeation enhancement of buflomedil hydrochloride from niosomes.
Conclusion: Thus, it was concluded that niosomal vesicles represented to be an efficient and
stable vesicular carrier for transdermal delivery of buflomedil hydrochloride.

Introduction investigated, niosomal vesicles have been found to be capable


enough as a transdermal drug carrier in enhancing the
Transdermal drug delivery system is considered to be an
permeation. These are the vesicular carriers prepared using
attractive alternative route to oral route of drug delivery
non-ionic surfactants and cholesterol (Prasanthi & Lakshmi,
(Prausnitz & Langer, 2008). This system uses skin for the
2012). Enhanced permeation through these vesicles is due
delivery of systemically acting drugs (Prasanthi & Lakshmi,
to the fact that these vesicles get interacted with stratum
2012). Despite of having the advantages of avoiding gastro-
corneum by aggregation, fusion and adhered to the cell surface
intestinal absorption of drugs, bypassing first-pass hepatic
causing a high-thermodynamic activity gradient of the drug
metabolism, controlling drug release (Gaikwad, 2013), non-
at the surface of vesicle and stratum corneum. This acts
invasive, self-administration (Prausnitz & Langer, 2008) and
as a driving force for the penetration of drugs across
improved patient compliance, this system has the limitation of
the stratum corneum. Niosomes also modify the structure of
slow diffusion of drugs due to lesser permeability across the
stratum corneum; loosen the intercellular lipid barrier of
skin. Thus, limiting the delivery of the drug through the skin
stratum corneum. Thereby, increasing the stratum corneum
(Gaikwad, 2013). Therefore, several approaches have been
permeability. Non-ionic surfactant present in niosomes works
used to weaken this barrier and to enhance the transdermal
as a permeation enhancer and plays a key role in enhancing the
delivery of the drug into the systemic circulation. One of these
permeation (Rahimpour & Hamishehkar, 2012). Buflomedil
approaches is the use of vesicle-based formulations as
hydrochloride [4-(1-pyrrolidinyl)-1-2,4,6-trimethoxyphenyl)-
transdermal system. Of the various vesicular systems
1-butanone hydrochloride] is a vasoactive drug employed
widely in the treatment of vascular diseases increasing the
Address for correspondence: Nida Akhtar, Assistant Professor, peripheral and arterial blood flow in ischaemic patients (El-
School of Pharmaceutical Sciences, IFTM University, Lodhipur-
Rajput, Moradabad 244102, Uttar Pradesh, India. E-mail:
Desoky et al., 2010). It is also employed in the treatment of
nidakhtr378@gmail.com dementia, disorientation, insomnia, intermittent claudication,
2 S. Arkvanshi et al. J Liposome Res, Early Online: 1–11

ischaemic rest pain, loss of concentration, loss of memory, Selection of concentration of cholesterol
Raynaud’s syndrome, etc. The present investigation was
Blank niosomes were prepared using different concentration
undertaken to develop niosomal formulations that can improve
of cholesterol to select the optimum concentration required
the transdermal delivery of buflomedil hydrochloride. As
for the preparation of niosomes. For this, niosomes were
postulated by various authors, the oral and parenteral forms
prepared at concentrations 0.1–5.0% w/v. Niosomal suspen-
administered of buflomedil hydrochloride has risk of adverse
sions were then visualized by taking micrographs through
reactions and may cause premature inactivation of the drug by
optical photomicrograph (Photomicrograph, HICON, Delhi,
first pass hepatic metabolism. Liposomes were then formulated
India) at 100.
by Roesken et al. to overcome the limitations possessed by oral
and parenteral formulations. But, these vesicles were found to
Preparation of buflomedil hydrochloride niosomes
possessed limited permeability across the skin (Roesken et al.,
2000). Thus, in this research work, an attempt has been made to Niosomes loaded with buflomedil hydrochloride were for-
formulate niosomal-based transdermal delivery system encap- mulated by ether injection method based on 32 factorial design
(Table 1). 32 Factorial design is a three-level and two factors-
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14

sulating buflomedil hydrochloride, a vasoactive drug. This can


improve the therapeutic efficacy of drug due to its greater based factorial design as reported by Webb (1971). In total,
concentration and longer time duration at the desired site of there are nine runs or combinations. In designing niosomes,
action with improved permeability across the skin. It also cholesterol and span 60 were selected as two independent
avoids inactivation of drug as it bypasses the first-pass hepatic variables, both at three different levels (1, 0, +1). In the
metabolism. Further, it can also reduce the side effects design, the lower concentration of cholesterol, i.e. 1.0%w/v,
associated with the drug usage (Muri et al., 2010). In vitro has been assigned at 1 level, whereas 2.0% w/v concentration
permeability study was done to layout the comparison among was assigned the level 0. The concentration at highest level +1
control formulation, niosomes and reference liposomes for was 3.0% w/v. In case of span 60, concentrations at three levels
improved permeability across the skin. from lowest to highest (1, 0, +1) were 0.5, 1.0 and 3.0% w/v,
respectively. Based on this factorial design, nine niosomal
Materials formulations were developed. Span 60, cholesterol and drug
were dissolved in a mixture of diethyl ether and methanol as
Buflomedil hydrochloride was obtained as a gift sample from
per the composition given in Table 1 and was stirred
Fresenius Kabi Oncology Ltd. (Kalyani, West Bengal, India).
For personal use only.

continuously on a magnetic stirrer (Macro Scientific Works,


Cholesterol, Span 40, Span 60 and Span 80 were obtained
Delhi). Distilled water was heated to 55 ± 1  C. Organic phase
from Central Drug House (CDH) Pvt. Ltd. (New Delhi,
was then injected into an aqueous phase.
India). Methanol and sodium chloride extrapure LR were
procured from CDH Pvt. Ltd. (New Delhi, India). Potassium
Purification of drug-loaded niosomes
dihydrogen orthophosphate was purchased from SD Fine
chemicals Ltd. (Mumbai, India). HIMEDIA Dialysis mem- Drug-loaded niosomes were purified by dialysis membrane
brane 150 was procured from Sigma Aldrich (Mumbai, India). technique to remove the free drug from niosomal suspension.
Diethyl ether, hydroxy propyl methyl cellulose (HPMC K4M) For this, Hi-media dialysis membrane was kept in saline
and sodium chloride were purchased from Central Drug solution for 2 h before dialysis to ensure complete wetting of
House (CDH) Pvt. Ltd. (New Delhi, India). Sodium hydroxide the membrane. Niosomal vesicles loaded with buflomedil
pellets LR were procured from RFCL Ltd. (New Delhi, hydrochloride were placed in a dialysis bag, which was
India). Glycerin was procured from SD Fine Chemicals Pvt. transferred into 200 ml of phosphate buffer pH 7.4. The
Ltd. (Mumbai, India). receiver medium was stirred with a magnetic stirrer at 500 rpm

Methods Table 1. 32 Factorial design of liposomal and niosomal formulations of


buflomedil hydrochloride.
Preliminary studies on blank niosomes
Preliminary studies were performed to optimize the blank Formulation Drug Cholesterol Span 60 Diethyl ether Methanol
code (mg) (% w/v) (% w/v) (% w/v) (% w/v)
niosomes prepared by ether injection method described later.
Blank niosomes were prepared using different types of Span Niosomes
F1 10 1.0 () 0.5 () 1.0 1.0
(Span 40, 60 and 80). An appropriate type of span and its F2 10 1.0 () 1.0 (0) 1.0 1.0
optimum concentration were selected. Selection of required F3 10 1.0 () 3.0 (+) 1.0 1.0
concentration of cholesterol was also done by preparing the F4 10 2.0 (0) 0.5 () 1.0 1.0
blank niosomes. F5 10 2.0 (0) 1.0 (0) 1.0 1.0
F6 10 2.0 (0) 3.0 (+) 1.0 1.0
F7 10 3.0 (+) 0.5 () 1.0 1.0
Selection of type and concentration of span F8 10 3.0 (+) 1.0 (0) 1.0 1.0
F9 10 3.0 (+) 3.0 (+) 1.0 1.0
Blank niosomes containing span 40, 60 and 80 were prepared. Liposomes
For this, niosomal suspensions were visualized by taking F10 10 1.0 () – 1.0 1.0
micrographs through optical photomicrograph (Photomicro- F11 10 2.0 (0) – 1.0 1.0
graph, HICON, Delhi, India) at 100. Further, blank niosomes F12 10 3.0 (+) – 1.0 1.0
Fx* 10 1.5 1.5 1.0 1.0
were prepared by changing the concentration of span 60 in the
range of 0.1–3.0% w/v to optimize its concentration. *Extra design check point formulation.
DOI: 10.3109/08982104.2014.974058 Buflomedil hydrochloride niosomal patch for transdermal delivery 3

(13.98  g). Five millilitres of sample was withdrawn at liposomal vesicles (F12) were placed in a dialysis bag. The
appropriate time intervals and replaced with an equal volume dialysis bag was then placed in 250 ml of phosphate buffer pH
of fresh media and analyzed spectrophotometerically for the 7.4 in a beaker with constant stirring at 37  C for 8 h. Samples
amount of free drug. Purification time was optimized by withdrawn at appropriate time intervals were analyzed
applying statistical paired t test at 5% level of significance. spectrophotometerically. Percent cumulative amount of drug
released was then determined.
Evaluation of niosomes
Stability study
Vesicle size and zeta potential
Stability of the optimized (F9) and reference (F12) formula-
Vesicle size, zeta potential and polydispersity index were tion was determined by storing the vesicles at 4  C
measured by Zetasizer ver. 6.01 (Malvern Instruments Ltd, (refrigerated temperature) and 25  C (room temperature) for
Malvern, Worcestershire) by diluting one drop of liposomal 6 months. After appropriate time intervals, samples were
and niosomal suspension with hydroethanolic solution at withdrawn and evaluated for their mean particle size, zeta
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14

25  C in clear disposable zeta cells. All the measurements potential and percent entrapment efficiency.
were done in triplicate for each sample.
Preparation of patches
Entrapment efficiency
Solvent evaporation method
Purified vesicular suspension was transferred into the centri-
fuge tube and centrifuged (Macro Scientific Works, Transdermal patches loaded with niosomal vesicles were
New Delhi, India) for 1 h at 4000 rpm (894.0  g). The prepared using solvent evaporation method. HPMC K4M was
sediment was lysed using methanol and filtered through nylon dissolved in a mixture of chloroform and methanol (6:4).
filter disc (0.22 mm). The drug was assayed both in the Optimized niosomes and reference liposomal formulation(s)
sediment and supernatant to determine the entrapment were centrifuged at 2000 rpm (447.0 g), and the pellets were
efficiency by the following equation: incorporated in the above polymer base. Glycerin was added
under continuous stirring on the magnetic stirrer (Macro
% Entrapment efficiency ¼ ½Wa  ðWs þ WP Þ=Wa , ð1Þ Scientific Works, for 5–6 h. The mixture was spread in the
petridishes. The petridishes were then placed in hot air oven
For personal use only.

where Wa is the amount of drug added in the system, Ws is the


amount of drug in the supernatant after centrifugation and WP at 36  C temperature.
is the amount of drug in the purification medium.
Evaluation of patches
Validation of experimental design Differential scanning calorimetry
The polynomial equation was generated for dependent Differential scanning calorimetric analysis of drug, span 60,
variable using Design Expert Software version 8.0.5 (Stat- cholesterol, HPMC K4M (Hydroxy Propyl Methyl Cellulose),
Ease Inc., Minneapolis, MN). An extra design check point niosomal patch, liposomal patch and control patch were
formulation (fx) was developed by selecting the levels as 1.5% carried out. The analysis was conducted in the heating
w/v of cholesterol and 1.5% w/v of span 60. This was used to range of 54–260  C in nitrogen atmosphere at the rate of
validate the obtained polynomial equation model. A statistical 150 ml/min), using differential scanning calorimetry (DSC,
model consisting of interactive and polynomial terms was Pyris Diamond TG/DTA, PerkinElmer, Singapore).
used to evaluate the responses. Polynomial and transformed
polynomial equations were obtained. Extra design check point Scanning electron microscopy
formulation was then evaluated for dependent variable, i.e.
percent entrapment efficiency to get the experimental value. Scanning electron microscope (Model JSM 6360, Jeol Make,
This value was compared with the predicted value obtained United Kingdom) was used to characterize the loading of
from transformed polynomial equation and evaluated statis- niosomes in patches and reference liposomal patches as well
tically by pooled t test at 5% level of significance. as control patches. The samples were visualized under
scanning electron microscope at 1500 magnification with
accelerating voltage of 17 kV.
Evaluation of optimized niosomes
Scanning electron microscopy Thickness
Scanning electron microscope (Model JSM 6360, Jeol Make, Thickness was measured using screw gauge (Sterling
Tokyo, Japan) was used to characterize optimized niosomes Manufacturing Company, India). The study was done in
and reference liposomal formulation. The samples were triplicate.
visualized under scanning electron microscope at 1500
magnification with accelerating voltage of 17 kV. Weight uniformity
The patches were subjected to weight variation test by
In vitro release study
weighing the selected patches individually. Three dried
For in vitro release study, dialysis membrane was washed and patches were weighed on an electronic balance (Citizen
soaked in saline solution. Optimized niosomal (F9) and Scale CY220). The study was done in triplicate.
4 S. Arkvanshi et al. J Liposome Res, Early Online: 1–11

Tensile strength 32  C ± 0.5  C was stirred using magnetic stirrer at 60 rpm.


One-millilitre sample was withdrawn from the receptor
In order to determine the elongation as a tensile strength, the
compartment at appropriate time intervals and analyzed
polymeric patch was pulled by means of a pulley system;
spectrophotometerically. An equal volume of fresh phosphate
weights were gradually added to the pan to increase the
buffer pH 7.4 was replaced after each sampling into the
pulling force till the patch was broken. The elongation, i.e. the
compartment (Wahid et al., 2008). The study was performed
distance travelled by the pointer before break of the patch,
in triplicate, and average values were calculated. The percent
was noted with the help of magnifying glass on the graph
cumulative amount of drug permeated across the skin per
paper. Equation 2 was used to calculate the tensile strength.
square surface area was evaluated and plotted against time to
Tensile load at break calculate the transdermal flux.
Tensile strength ¼ ð2Þ
Dis tan ce travelled by broken patch
Results and discussion
Flatness Preliminary studies on blank niosomes
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14

Three longitudinal strips were cut out from each patch, i.e. Blank niosomes were formulated by ether injection method,
one from the centre, one from the left side and third one and several preliminary optimization studies were conducted
from the right side. The length of each strip was measured. in order to optimize the methodology. From the micrographs
The change in length because of non-uniformity in flat- taken, it was observed that in case of span 40, vesicles formed
ness was measured by determining percent constriction, were non-uniform in size and had indefinite structure, while
with 0% constriction equivalent to 100% flatness (Patel et al., span 80 did not accompanied the formation of vesicles.
2009). However, niosomes formulated with span 60 were uniform
and spherical in shape with well defined and specific
Folding endurance boundary (Figure 1). The results obtained are in accordance
This was determined by repeatedly folding one film at the with the fact that span 60 is the better surfactant of all the
same place till it broke. The number of times the film could surfactants as it is having high-phase transition temperature
be folded at the same place without breaking/cracking gave and possesses low-hydrophilic–lipophilic balance (HLB) so it
the value of folding endurance. form the vesicles of appropriate size (Akhilesh et al., 2012).
For personal use only.

Critical packing factor (CPF) is another parameter considered


Percentage of moisture content behind the selection of span 60. It is the molecular charac-
teristic of surfactants that depicts whether the aggregate form
The patches were weighed individually and kept in desicca- is spherical micelles, inverted micelles or vesicles
tors containing activated silica at room temperature for 24 h. (Israelachvili, 1994). Surfactants with CPP value 0.5–1 can
Individual patch was weighed repeatedly until they showed a only form vesicles. As span 60 was reported to have above
constant weight. The percentage of moisture content was specified CPP value can lead to the formation of vesicles
calculated as the difference between initial and final weight (Akhilesh et al., 2012). Span 60 has relatively high hydro-
with respect to final weight. phobicity and less value of critical packing parameter. So, on
adding cholesterol, it has potential to form optimum mem-
Determination of percent drug content brane curvature for lamellar (vesicular) structure (Manosroi
Drug content was determined by taking the patches of 1 cm2 et al., 2003). Thus, span 60 was selected for preparing
surface area and dissolved in 5 ml methanol. Vortex shaking niosomes. Further, an appropriate concentration of span 60
(Vortex shaker, HICON, India) was done for 5 min, and was selected, using varying concentrations of it. It was
volume was made up to 10 ml with methanol and examined observed that at 0.5% w/v concentration of span 60, niosomal
spectrophotometrically (Shimadzu UV 1800, Japan) for drug vesicles formed were firm, with well-defined boundary and
content. spherical in shape confirming uniformity. At concentration
below 0.5% w/v, vesicle could not be formed and on
In vitro release studies increasing the concentration of span 60 beyond 0.5% w/v
till 5.0% w/v, the size of vesicle increased with loss in
In vitro release studies were performed for niosomal patches, uniformity in size. Thus, the optimum concentration of span
liposomal and control patches. For this study, Franz Diffusion 60 was identified to be 0.5, 1.0 and 3.0% w/v. After the
cell (Bhanu Scientific Instrument CO, Bangalore, India) was selection of span and its concentration, optimum concentra-
used with a receptor compartment of 10 ml capacity and area tion of cholesterol was selected. Among the concentration of
3.14 cm2. Hi-media dialysis membrane 150 (molecular weight cholesterol taken (0.5–5.0% w/v), it was observed that
cut off (MWCO) 12–14 kDa; Prajapati et al., 2011a) was uniformity of vesicles was good at concentration 1.0–3.0%
soaked in phosphate buffer pH 7.4 and mounted on the Franz w/v. This also guided the selection of levels of span 60 and
diffusion cell. The transdermal patch of area 3.14 cm2 area cholesterol for 32 experimental design.
was placed in the donor compartment and it was separated
from the receptor compartment by dialysis membrane. The
Optimization of purification time
donor and receptor compartment was joined together using
clamp (Prajapati et al., 2011b). The receptor compartment Selected formulation (F9) was subjected to purification, and
containing 10 ml of the phosphate buffer pH 7.4 maintained at the amount of free drug released was observed for 60 min.
DOI: 10.3109/08982104.2014.974058 Buflomedil hydrochloride niosomal patch for transdermal delivery 5
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14

Figure 1. Photomicrographs of niosomes prepared with (a) Span 40; (b) Span 60 and (c) Span 80 at 100.

Initially, the amount of free drug released increased till 20 min Table 2. Pharmacotechnical properties of niosomal vesicles (F1–F9) and
and reached plateau level by 30 min (data not shown). Paired t reference liposomal vesicles (F10–F12).
test was applied to determine any significant difference in the
amount of free drug released at different time intervals. It was Zeta Percent
For personal use only.

Formulation Particle size Polydispersity potential entrapment


observed that there was a significant difference (p50.05) in code range (mm) index (mV) efficiency
the amount of free drug released between 10 and 30 min, but
there was no significant difference (p40.05) in amount of F1 2.87 ± 1.9 0.312 ± 0.02 61.3 ± 0.42 65.05 ± 0.14
F2 2.95 ± 0.5 0.234 ± 0.11 65.1 ± 0.58 72.11 ± 0.11
free drug released after 30 and 40 min. Therefore, all the F3 3.77 ± 2.4 0.136 ± 0.14 65.5 ± 0.61 78.26 ± 0.09
formulations were subjected to purification by dialysis F4 4.08 ± 2.1 0.245 ± 0.22 69.4 ± 0.54 81.24 ± 0.21
technique for a period of 30 min. F5 5.16 ± 1.1 0.218 ± 0.10 72.7 ± 0.23 83.31 ± 0.05
F6 6.43 ± 2.3 0.308 ± 0.15 74.8 ± 0.39 85.57 ± 0.16
F7 7.58 ± 1.4 0.276 ± 0.21 79.2 ± 0.70 89.08 ± 0.16
Evaluation of niosomes F8 8.73 ± 2.7 0.225 ± 0.03 81.9 ± 0.37 93.44 ± 0.13
F9 10.09 ± 1.2 0.121 ± 0.07 85.4 ± 0.56 97.09 ± 0.11
Determination of vesicle size and zeta potential F10 3.98 ± 2.2 0.309 ± 0.18 57.4 ± 0.33 56.77 ± 0.03
F11 7.12 ± 1.8 0.248 ± 0.19 62.9 ± 0.59 61.46 ± 0.22
Niosomes and reference liposomal formulations were eval-
F12 13.99 ± 1.6 0.201 ± 0.12 67.5 ± 0.47 64.12 ± 0.10
uated for particle size, polydispersity index and zeta potential.
It was observed that the niosomal vesicles of
formulations (F1–F9) varied in size range of 2.87 ± 1.9 mm
to 10.09 ± 1.2 mm in size where as reference liposomal vesicles of particles within the formulation, whereas if it is greater than
(F10–F12) ranged between 2.54 ± 2.2 mm and 7.12 ± 1.6 mm 0.3, it dictates the heterogeneous nature of the particle
(Table 2). It was observed in niosomes that as the concentra- distribution (Maurya et al., 2010). It was found to be minimum
tion of span 60 increased, vesicular size of the particles also for F9 0.121 ± 0.07 among niosomal vesicles and
increased. Further it has also been evaluated that with 0.201 ± 0.120 (F12) among reference liposomal formulations
enhanced concentration of cholesterol, vesicle size also showing the homogeneous nature of the formulations. F9 and
increased. Availability of higher amount of raw materials F12 made with highest levels of cholesterol and span displayed
probably led to the formation of larger-sized vesicles (Zhaowu least PDI probably due to dominant surfactant action of span
et al., 2009). Similarly, in case of liposomal formulations, 60 (Akhilesh et al., 2012). Zeta potential is another parameter
vesicle size was dependent primarily on the amount of responsible for the thermodynamic stability. Niosomal ves-
cholesterol, as the concentration of cholesterol increases, icles had higher zeta potential 85.4 ± 0.56 mV as compared
the size of vesicles increased from formulation F10 to F12. to reference liposomal formulations, i.e. 67.5 ± 0.47 mV.
The results are found to be consistent with the earlier reports The negative charge on the vesicles might be the charge due to
made by Kamboj et al. where the authors reported an increase the drug. As buflomedil hydrochloride carries negative charge
in niosomal vesicular size on increasing cholesterol concen- concentrated on the Cl (chloride) atom and methoxy group
tration (Kamboj et al., 2014). The polydispersity index (PDI) (–OCH3) (Eicher et al., 2009). Cholesterol is electrically
specifies the distribution of vesicles as it defines the meas- neutral (Masterjohn, 2005). Span 60 also carries neutral charge
urement of homogeneity of a vesicular formulation. A PDI being a non-ionic surfactant (Akhilesh et al., 2012). So, the net
value of less than 0.3 indicates the homogeneous distribution charge on the vesicles may due to the drug. Analysis of zeta
6 S. Arkvanshi et al. J Liposome Res, Early Online: 1–11

potential values indicates higher magnitude of negative charge


on niosomal vesicles.

Entrapment efficiency
Among all the niosomal formulations, percent entrapment
efficiency (Table 2) was found to be maximum in formulation
F9 (97.09% ± 0.11%), where as in case of liposomes,
formulation F12 showed maximum percent entrapment effi-
ciency of 64.12% ± 0.10%. Thus, it was observed that
niosomes possessed higher drug entrapment as compared to
liposomes. The presence of span 60 had a positive impact on
percent drug entrapment of the vesicular carrier. The
percentage entrapment efficiency was found to be increased
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14

by increasing concentration of cholesterol. At a ratio of 1:1 of


span 60:cholesterol, entrapment efficiency was found to be
maximum (Kamboj et al., 2014). Figure 2. Response surface plot showing the effect of levels of
cholesterol and Span 60 on entrapment efficiency.
Validation of experimental design
done by scanning electron microscopy (SEM). SEM images,
The 32 factorial design was validated using Design Expert
in both cases, depicted the presence of almost uniform,
Software version 8.0.5 (Stat-Ease). A general statistical model
spherical-shaped vesicles and absence of aggregates
can be developed with respect to the data obtained from the
(Figure 3). However, niosomal vesicles more uniform as
formulations subjected to optimization. The model depicted
compared to liposomal vesicles.
can be characterized using the polynomial equation repre-
senting the respective response data. This can be given as In vitro release study
follows:
In vitro release from the niosomes compared to the liposome
% Entrapment efficiency formulation was determined. Percent cumulative drug release
For personal use only.

¼ þ85:14  8:16x1  1:45x2  4:29x21 þ 0:72x22 ð3Þ in case of niosomes and liposomes was observed to be
97.21 ± 1.21 and 63.78 ± 1.34, respectively, after 8 h (data not
þ 1:24x1 x21 þ 2:36x2 x21  1:18x1 x22  1:35x2 x22
shown). It was found that the drug release from niosomal
whereas the final transformed equation, Equation (2), vesicles was more as compared to liposomes. This was due to
obtained by removing all the insignificant values was higher percent of drug entrapped in them, which increases the
cumulative amount of drug release.
% Entrapment efficiency ¼ þ85:14  8:61x1 ð4Þ
From the above polynomial equations, response surface Stability studies
plot was generated which was then used to predict the Stability studies were carried out by storing the samples at
response of dependent variables at the intermediate levels of 4  C and 25  C to investigate the long-term stability of the
independent variables. From the response surface plot vesicles at its storage condition. The stability data had shown
(Figure 2), it was observed that as the levels of both in Table 3. As indicated, there was no significant difference in
cholesterol and span 60 increased, entrapment efficiency vesicular size, zeta potential and percent entrapment effi-
was also increased. The percent entrapment efficiency of the ciency of niosomes before and after storage at refrigerated
extra design check point formulation (Fx) was found to be temperature. This dictated good stability of niosomal vesicles
87.62 ± 7.58%. Comparison with predicted value of 85.14% upon storage because of the combinatorial stable effect
obtained from the transformed polynomial equation indicated exerted by the presence of both cholesterol and span 60. As
no significant difference (p40.05) between the two values addition of higher concentration of cholesterol provides
when checked using pooled t test thus validating the design. stability to the vesicles (Das & Palei, 2011) and aggregation
of vesicles can be prevented by the addition of an amphiphile
Selection of optimized formulation (span 60). Thereby, stabilizing the system by repulsive steric
The optimized formulation of niosomal formulation was or electrostatic effects (Akhilesh et al., 2012). However, at
identified as F9 on the basis of highest entrapment efficiency higher temperature (room temperature 25  C), significant
of 97.09% ± 0.11%. Similarly, the liposomal formulation F12 difference in vesicular size, zeta potential and percent
(reference) was selected on the basis of maximum entrapment entrapment efficiency of niosomes before and after storage
efficiency of 64.12% ± 0.10%. was observed. Vesicles may tend to aggregate and form
vesicles of larger size. Thereby, vesicular size of vesicles was
Evaluation of optimized niosomes found to be increased. Aggregation of these vesicles may
cause drug leakage, decreasing their entrapment efficiency.
Vesicular characterization by scanning electron microscopy Thus, it was concluded that storage under refrigerated
Vesicular characterization of optimized niosomes (F9) and conditions showed greater stability as compared to the room
liposomes (F12) loaded with buflomedil hydrochloride was temperature (Sakthivel et al., 2012).
DOI: 10.3109/08982104.2014.974058 Buflomedil hydrochloride niosomal patch for transdermal delivery 7
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14

Figure 3. Scanning electron microscopic images of (a) niosome (F9) and (b) Liposome (F12).

Table 3. Stability study data of the optimized buflomedil hydrochloride loaded niosomes (F9) and reference liposomes (F12) at refrigerated (4  C) and
room temperature (25  C).

Parameters

Formulation code Temperature ( C) Time period (Months) Vesicle size (mm) Zeta potential (mV) % Entrapment efficiency
F9 4 (refrigerated temperature) 0 10.09 ± 1.2 85.4 ± 0.56 97.09 ± 0.11
6 23.11 ± 3.4 71.9 ± 0.26 88.15 ± 0.08
F12 0 7.12 ± 1.6 67.5 ± 0.47 64.12 ± 0.10
6 35.88 ± 4.2 56.2 ± 0.35 53.16 ± 0.17
F9 25 (room temperature) 0 10.09 ± 1.2 85.4 ± 0.56 97.09 ± 0.11
6 47.33 ± 2.8 56.9 ± 1.10 66.75 ± 0.19
For personal use only.

F12 0 7.12 ± 1.6 67.5 ± 0.47 64.12 ± 0.10


6 65.14 ± 1.9 42.4 ± 0.73 45.31 ± 0.23

Evaluation of niosomal patches loaded with cholesterol) were of optimum suitability because of their
buflomedil hydrochloride compatibility with the drug (Verma & Chandak, 2009).
Differential scanning calorimetry
Scanning electron microscopy of patches
Differential scanning calorimetric (DSC) studies were con-
Scanning Electron microscopic images of niosomal and
ducted for drug, niosomal patch and liposomal patch. DSC
liposomal patches highlighted the loading of vesicles within
thermograms of pure drug, span 60, cholesterol and HPMC
the dermatological patch system. However, the distribution
K4M were shown in Figure 4. DSC thermograms of niosomal
of niosomes in patches was more uniform as compared to
patch, liposomal and control patch were shown in Figure 5.
liposomal vesicles (Figure 6).
It was observed that buflomedil hydrochloride (BH) exhibited
a sharp endothermic peak at 196.89  C, showing melting of
Pharmacotechnical properties of patches
pure drug. The melting point of buflomedil hydrochloride was
reported to be 195–198  C (Clissold et al., 1987). Span 60 and The pharmacotechnical properties of patches were given in
cholesterol showed a sharp endothermic peak at 56.25 and Table 4. Thickness was found to be 0.30 ± 0.48 mm in case of
148  C, respectively. These two peaks showing the melting niosomal patches, whereas liposomes possessed the thickness
point of both the excipients, i.e. 54–57  C (Peltonen, 2001) of about 0.093 ± 0.31 mm. Thinner the patches, more flexible
and 148–150  C (Ohvo-Rekila et al., 2002). Pure HPMC K4M it is (Mamatha et al., 2010). Niosomal patches
exhibited the slight broad peak at temperature range of (4.83 ± 1.79 kg/cm2) showed more tensile strength as com-
50–66.5  C. In case of both liposomal and niosomal patches, a pared to liposomal patches (4.30 ± 1.69 kg/cm2). It was
sharp endothermic peak of pure drug (BH) was disappeared. evaluated from the results that niosomes are more strong
A broad endothermic peak was observed in between 50 and and flexible as compared to liposomal patches (Rama &
87.5  C. This may be attributed due to the dispersion of drug Shantha, 2012). Percent moisture content was found to be less
in the polymeric matrix (Bhosale et al., 2011). No other sharp for niosomal patches, i.e. 6.0 ± 2.10%. Low-moisture content
endothermic peak was observed, indicating the fact that no helps the formulation to remain stable and make them free
interaction took place between drug, span 60, cholesterol and from being a brittle and dry film. Percent flatness was found
polymer (HPMC K4M). Thus, from this study, it was to be similar in both niosomal as well as liposomal patches
concluded that for the preparation of liposomal and niosomal (100% for both). Flatness study revealed that the formulation
patches, polymer and all other excipients (span 60, possess same length before and after the test. Thus, from this
8 S. Arkvanshi et al. J Liposome Res, Early Online: 1–11
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14
For personal use only.

Figure 4. DSC thermogram of (a) pure drug; (b) cholesterol; (c) Span 60 and (d) HPMC K4M.

Figure 5. DSC thermogram of (a) control patch; (b) niosomal patch and (c) liposomal patch.

test, it was concluded that there was no constriction in patches


In vitro release studies
and had smooth and flat surface. It was expected that the
smoothness would be retained even after the application onto In vitro permeability studies were conducted for liposomal
the skin. Folding endurance was found to be 209 ± 1.8 which and niosomal patch using dialysis membrane in phosphate
showed that folding endurance was greater than liposomal buffer pH 7.4 up to 8 h (Liu et al., 2011). The in vitro release
patches. This parameter specifies the fact that integrity of the parameters had shown in Table 5. Cumulative release profiles
patches was maintained over the skin (Mamatha et al., 2010). of niosomal patches, liposomal and control patches were
Percent drug content was found to be maximum in case compared (Figure 7). Regardless of whether transdermal
of niosomal patches (97.09 ± 8.04) due to maximum absorption has a systemic action or a local effect, a drug
drug entrapment possessed by the niosomes due to max- should pass through the stratum corneum. However, due to
imum concentration of cholesterol and span 60. As span barrier effect of the stratum corneum, many drugs cannot play
60 due to low-HLB value provided higher drug loading a full role (Liu et al., 2011). Flux, release rate and release
(Dharashivkar et al., 2014). coefficient were found to be higher in case of niosomal
DOI: 10.3109/08982104.2014.974058 Buflomedil hydrochloride niosomal patch for transdermal delivery 9
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14
For personal use only.

Figure 6. Scanning electron microscopic images of (a) control patch; (b) niosomal patch and (c) liposomal patch.

Table 4. Pharmacotechnical properties of niosomal loaded patches and reference liposomal patches.

Formulation code Niosomal patch (NP) Liposomal patch (LP) Control patch (CP)
Parameters
Thickness (mm) 0.30 ± 0.48 0.091 ± 0.31 0.085 ± 0.24
Weight variation (mg/cm2) 0.483 ± 0.57 0.325 ± 0.47 0.603 ± 0.63
Flatness (%) 99.98 ± 0.10 97.12 ± 1.11 94.68 ± 0.55
Folding endurance 209 ± 1.80 196 ± 1.43 199 ± 1.51
Tensile strength (kg/cm2) 4.83 ± 1.79 4.30 ± 1.69 3.6 ± 1.55
Moisture content (%) 6.66 ± 2.10 7.76 ± 2.44 9.0 ± 2.26
Drug content (%) 97.09 ± 1.11 75.12 ± 1.53 71.33 ± 1.11

patches as compared to liposomal patches and control value of both liposomal patch and niosomal patch was less
patches. Surfactant present in niosomes act as a penetration than 50 (Table 5) which implies dissimilarity among release
enhancer which contribute in the permeation enhancement of profiles from control patch. Also, with reference to liposomal
buflomedil hydrochloride from niosomes. Further, niosomes patch (LP), niosomal patch release profile showed higher
undergo fusion at the interface of stratum corneum and high- dissimilarity (f2 ¼ 39) between two profiles. The curve was
drug concentration in bilayers leads to improved thermo- fitted by zero order, first order, Higuchi, Korsmeyer–Peppas
dynamic activity of buflomedil hydrochloride in the stratum and Hixon–Crowell release kinetic model. The order for the
corneum. These results are in accordance with the report release was chosen to investigate the possible mechanism of
evaluated by Fang et al., in which it was described that drug release and release rate as indicated by higher
niosomal formulations was superior to liposomal and control determination coefficient (r2) (Sakthivel et al., 2012). The
formulations in enhancing the release across the skin (Fang coefficient of determination (r2) in case of zero and first order
et al., 2011). The percent cumulative amount of drug released was found to be 0.9906 and 0.9019, respectively, in case of
was found to be 95.12 ± 1.12 in case of niosomal patches, niosomal patches (Table 6). As r2 value was more in zero
which greatly enhances the effect of buflomedil hydrochlor- order, specifying zero order controlled release pattern.
ide. On comparing the release profiles with that of control Further, in order to determine whether diffusion is involved
patch profile (reference) using one way ANOVA, significant in the drug release, the data were subjected to Higuchi model.
difference (p50.05) in permeability was observed. Similarity The lines obtained were comparatively linear with r2 value of
(f2) and dissimilarity factor (f1) were also calculated. The f2 0.9994 indicating that the diffusion may be the mechanism
10 S. Arkvanshi et al. J Liposome Res, Early Online: 1–11

Table 5. In vitro release study of buflomedil hydrochloride loaded niosomal patch and liposomal patch across the dialysis membrane.

Formulation % Cumulative drug Flux Release rate Release coefficient


code released (8 h) (mg/cm2/min) (mg/cm2/min0.5) (cm/min  102) ER f2
NP 95.12 ± 1.19 16.67 ± 1.53 57.80 ± 1.15 0.531 ± 1.50 2.62 30
LP 82.21 ± 1.24 8.83 ± 1.28 45.62 ± 1.70 0.265 ± 1.440 2.07 49
Control patch* 70.10 ± 1.33 1.04 ± 1.31 22.05 ± 1.01 0.033 ± 1.24 1.00 100

ER Enhancement ratio.
*Reference formulation for calculation of f2

Thus, niosomal system signifies to be a better carrier for


effective and safe transdermal drug delivery.
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14

Acknowledgements
The authors are thankful to IFTM University, Moradabad, for
providing all the facilities required for conducting the
research work.

Declaration of interest
Authors declared no conflict of interest.

References
Akhilesh D, Bini KB, Kamath JV. (2012). Review on span-60 based non-
Figure 7. Comparative in vitro release profile of buflomedil hydro- ionic surfactant vesicles (niosomes) as novel drug delivery. Int J Res
chloride from control, liposomal and niosomal patch. Pharm Biomed Sci 3:6–12.
Bhosale NR, Hardikar SR, Bhosale AV. (2011). Formulation and
For personal use only.

evaluation of transdermal patches of ropinirole hydrochloride. Res J


Table 6. ‘‘r2’’ Values of niosomal patch, liposomal patch and control Pharm Biol Chem Sci 2:138–80.
patch showing various kinetic models. Clissold SP, Lynch S, Sorkin EM. (1987). Buflomedil: a review of its
pharmacodynamics and pharmacokinetic properties, and therapeutic
Coefficient of determination (r2) efficacy in peripheral and cerebral vascular diseases. Drugs 33:
430–60.
Niosomal Liposomal Control Das MK, Palei NN. (2011). Sorbitan ester niosomes for topical delivery
Kinetic models patch (NP) patch (LP) patch (CP) of rofecoxib. Ind J Exp Biol 49:438–45.
Zero order 0.9906 0.9978 0.9982 Dharashivkar S, Shasrabuddhe S, Saoji A. (2014). Silver sulfadiazine
First order 0.9019 0.9506 0.9560 niosomes: a novel sustained release once a day formulation for burn
Higuchi 0.9994 0.9985 0.9755 treatment. Int J Pharm Pharm Sci 6:611–16.
Korsmeyer–Peppas 0.9866 0.9810 0.9730 Eicher T, Hauptmann S, Specicher A. (2009). The chemistry of
Hixon–Crowell 0.9687 0.9759 0.9772 heterocycles: structures, reactions, synthesis and application. 3rd ed.
Weinheim: Wiley Publishers.
El-Desoky HS, Ghoneim MM, Abdel-Galeil MM. (2010). Quantification
of the vasoactive agent buflomedil hydrochloride in pharmaceutical
formulation and human serum by stripping voltametry and liquid
involved in drug release from niosomal patch. The data were
chromatography. Acta Chim Slov 57:332–40.
then subjected to Korsmeyer–Peppas model to further confirm Fang J-Y, Hong C-T, Chiu W-T, Wang Y-Y. (2011). Effect of liposomes
the mechanism of drug permeation. The release exponent and niosomes on skin permeation of enoxacin. Int J Pharm 219:61–72.
‘‘n’’ values were determined. Based on ‘‘n’’ value, it can be Gaikwad AK. (2013). Transdermal drug delivery system: formulation
aspects and evaluation. Comprehensive J Pharm Sci 1:1–10.
explained that incorporated drug was released by the anom- Israelachvili J. (1994). The science and applications of emulsions: an
alous (non-fickian) type of diffusion, involving swelling of overview. Colloids Surf A: Physiochem Eng Aspects 91:1–8.
the matrix, as is evident by the slope values ranging in Kamboj S, Saini V, Bala S. (2014). Formulation and characterization of
between 0.5 and 1. Thus, it was concluded that diffusion is the drug loaded non-ionic surfactant vesicles (niosomes) for oral
bioavailability enhancement. Sci World J 2014:1–8.
dominant mechanism of drug release following non-fickian Liu X, Liu H, Liu J, et al. (2011). Preparation of a ligustrazine
type of diffusion (Wahid et al., 2008). ethosome patch and its evaluation in vitro and in vivo. Int J Nanomed
6:241–7.
Mamatha T, Rao JV, Ramesh G. (2010). Development of matrix type
Conclusion transdermal patches of lercanidipine hydrochloride: physicochemical
and in vitro characterization. DARU J Faculty Pharm 18:9–16.
Niosomal formulations represented a stable and efficient
Manosroi A, Wongtrakul P, Manosroi J, et al. (2003). Characterization of
vesicular carrier for enhanced transdermal delivery of vesicles prepared with various non-ionic surfactants mixed with
buflomedil hydrochloride that exhibited higher entrapment cholesterol. Colloids Surf B: Biointerfaces 30:129–38.
efficiency and stability than liposomal vesicles. Transdermal Masterjohn C. (2005). Cholesterol and health. Cholesterol Biochem 1:
1–3.
flux of niosomal loaded patches was higher than the reference Maurya SD, Prajapati SK, Gupta AK, et al. (2010). Formulation
liposomal patch as well as control patch. In vitro permeation development and evaluation of ethosomes of stavudine. Ind J Pharm
studies confirmed the enhanced permeation of drug. Educ Res 44:102–8.
DOI: 10.3109/08982104.2014.974058 Buflomedil hydrochloride niosomal patch for transdermal delivery 11
Muri EMF, Sposito MMM, Metsavaht L. (2010). Pharmacology of Rahimpour Y, Hamishehkar H. (2012). Niosomes as carrier in dermal
vasoactive drugs. Acta Fisiatr 17:22–7. drug delivery. In: Demir A. ed. Recent advances in novel drug carrier
Ohvo-Rekila H, Ramstedt B, Leppimaki P, Slotte JP. (2002). Cholesterol systems. InTech- Open Science Open Minds, 141–64.
interactions with phospholipids in membranes. Prog Lipid Res 41: Rama B, Shantha A. (2012). Optimization of methotrexate transdermal
66–97. patches: effect of variables on in vitro, ex vivo permeation and flux.
Patel RP, Patel G, Baria A. (2009). Formulation and evaluation of Int J Pharm Sci Lett 2:53–9.
transdermal patch of aceclofenac. Int J Drug Deliv 1:41–51. Roesken F, Uhl E, Curri SB, et al. (2000). Acceleration of wound healing
Peltonen L. (2001). The interfacial behaviour of sorbitan surfactant by drug delivery via liposomes. Langenbecks Arch Surg 385:42–9.
monolayers and the bulk properties of these surfactants as function of Sakthivel M, Kannan K, Manavalan R, Senthamasai R. (2012).
temperature [Dissertation]. Biocentri Viikki Universitatis Formulation and in vitro evaluation of niosomes containing
Helsingiensis. oxcarbazepine. Int J Pharmacy Pharm Sci 4:563–7.
Prajapati RK, Mahajan HS, Surana SJ. (2011a). PLGA based Verma PRP, Chandak AR. (2009). Development of matrix controlled
mucoadhesive microspheres for nasal delivery: in vitro/ex vivo transdermal delivery system of pentazocine. Acta Pharm 59:171–86.
studies. Ind J Novel Drug Deliv 3:9–16. Wahid A, Sridhar BK, Shivakumar S. (2008). Preparation and evaluation
Prajapati ST, Patel CG, Patel CN. (2011b). Formulation and of transdermal drug delivery system of etoricoxib using modified
evaluation of transdermal patch of repaglinide. ISRN Pharm chitosan. Ind J Pharm Sci 70:455–60.
2011:1–9. Webb SR. (1971). Small incomplete factorial experiment design for two-
Journal of Liposome Research Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/18/14

Prasanthi D, Lakshmi PK. (2012). Vesicles-mechanism of transdermal and three-level factors. Technometric 13:243–6.
permeation: a review. Asian J Pharm Clin Res 5:18–25. Zhaowu Z, Xiaoli W, Yangde Z, Nianfeng L. (2009). Preparation of
Prausnitz MR, Langer R. (2008). Transdermal drug delivery. Nat matrine ethosomes, its percutaneous permeation in vitro and anti-
Biotechnol 26:1261–8. inflammatory activity in vivo in rats. J Liposome Res 19:155–62.
For personal use only.

You might also like