You are on page 1of 10

Journal of Microencapsulation

Micro and Nano Carriers

ISSN: 0265-2048 (Print) 1464-5246 (Online) Journal homepage: http://www.tandfonline.com/loi/imnc20

Novel in situ gelling ocular inserts for


voriconazole-loaded niosomes: design, invitro
characterisation and invivo evaluation of the
ocular irritation and drug pharmacokinetics

Marwa Hassan Shukr

To cite this article: Marwa Hassan Shukr (2016): Novel in situ gelling ocular inserts for
voriconazole-loaded niosomes: design, invitro characterisation and invivo evaluation
of the ocular irritation and drug pharmacokinetics, Journal of Microencapsulation, DOI:
10.3109/02652048.2015.1128489

To link to this article: http://dx.doi.org/10.3109/02652048.2015.1128489

Published online: 06 Jan 2016.

Submit your article to this journal

Article views: 14

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=imnc20

Download by: [Gazi University] Date: 24 January 2016, At: 05:48


JOURNAL OF MICROENCAPSULATION, 2016
http://dx.doi.org/10.3109/02652048.2015.1128489

RESEARCH ARTICLE

Novel in situ gelling ocular inserts for voriconazole-loaded niosomes: design,


in vitro characterisation and in vivo evaluation of the ocular irritation and
drug pharmacokinetics
Marwa Hassan Shukr
National Organization for Drug Control and Research, Cairo, Egypt

ABSTRACT ARTICLE HISTORY


This work aimed to develop voriconazole in situ gelling ocular inserts loaded with niosomal suspension. Received 1 May 2015
Niosomes and mixed niosomes were prepared using span 40 and span 60 with pluronic L64 and pluronic Revised 28 September 2015
F127. The entrapment efficiency percentages (EE%), mean vesicle size, polydispersity index (PI), zeta Accepted 9 November 2015
potential and in vitro drug release of these niosomes were evaluated. F3-mixed niosomes prepared with Published online
span 60 and pluronic L64 was selected, due to its highest EE; optimum vesicle size with smallest PdI and 31 December 2015
suitable release pattern of the drug (63% after 8 h). In situ ocular inserts were prepared using sodium
Downloaded by [Gazi University] at 05:48 24 January 2016

KEYWORDS
carboxymethylcellulose (CMC Na) and sodium alginate (ALG) and characterised for surface morphology, In situ gel; niosomes; ocular
surface pH, water uptake, mucoadhesion and in vitro release. ALG in situ ocular insert (S2) was selected for insert; voriconazole
further in vivo evaluation of the ocular irritation and drug pharmacokinetics in the aqueous humour of
rabbits eyes. S2 in situ gelling ocular insert was non-irritant and showed significantly (p50.01) higher Cmax,
delayed Tmax and increased bioavailability.

Introduction increasing corneal uptake. Other possible reasons are their better
spreading ability on the lipophilic corneal surface and favourable
Fungal keratitis is a serious corneal disease that can impair vision or
rheological properties. Locally applied niosomal drug formulations
lead to total blindness, if improperly treated (Liang et al., 2009).
controlled ocular delivery and protected against drug metabolism
Voriconazole (VCZ), a second-generation anti-fungal agent, pos-
by the enzymes present in the lachrymal fluid (Abdelbary and El-
sesses phenomenal characteristics like broad-spectrum activity,
gendy, 2008).
activity against resistant fungal species and acceptable tolerability
The clinical efficacy of antifungal drugs depends on the
(Wiebe and Karriker, 2005; Lat and Thompson, 2011). Studies
achievement of an optimal drug concentration in the ocular tissues
suggested an excellent efficacy of VCZ against several ocular
(Kaur and Kakkar, 2010), and as a result, frequent dosing is usually
mycoses following topical administration (Gupta et al., 2011; de Sa
required. To overcome the side effects of pulsed dosing, attempts
et al., 2015).
Topical ocular drug delivery is the preferred route for the have been made to combine the advantages of a solid, single unit
treatment of disorders of the ocular surface and anterior segment, dosage for ocular drug delivery by using in situ gelling inserts. The
such as keratitis, conjunctivitis, glaucoma and diabetic keratopathy application of solid structures in the eye has gained some interest
(Abdelkader et al., 2011a, 2011b; Khutoryanskaya et al., 2014). over the years (Ahmed and Aboul-Einien, 2007). The inserts
Recent studies focussed on delivering the antifungal drugs in prepared by lyophilisation or freeze-drying method that hydrate
sufficient therapeutic concentrations to the cornea via several drug and gel quickly when exposed to lachrymal fluid will serve as an
delivery systems like surfactant-based elastic vesicles (Kaur et al., alternative to conventional niosomal eye drops and allow for
2012), mucoadhesive nanoparticles (Bhatta et al., 2012), liposomes simpler administration in the hope of increasing patient
(Dai et al., 2013), in situ gels (Gratieri et al., 2011), nanosuspensions acceptability.
(Luschmann et al., 2013), solid lipid nanoparticles (Basaran et al., Ideally, the formulation should be able to control the drug
2010), nanostructured lipid carriers (Shen et al., 2010) and release, remain in contact with the front of the eye for extended
microemulsions (Kapoor and Chauhan, 2008). period of time and avoid foreign body sensation (Bertram et al.,
Over the last three decades, niosomes of different compositions 2010; Farid et al., 2013).
used for topical ophthalmic drug delivery have increased/prolonged The proper choice of polymers can allow mucoadhesion and
the therapeutic effect while minimising toxic effects (Abdelkader controlled drug release and due to dissolution of the gel, there
et al., 2013). Niosomes have been reported as possible approach to would be no need to remove the insert after it is depleted of drug
improve the low corneal penetration and bioavailability character- (Werner, 2003). Therefore, ocular inserts have high potential as
istics for many drugs. The corneal penetration enhancing effect of ocular drug delivery system.
niosomes could be attributed to many factors. Disrupting the tight The aim of the study was to develop a novel niosomal solid
junctions of the corneal epithelium is partly responsible for topical ocular drug delivery system. A number of non-ionic

CONTACT Marwa Hassan Shukr marwa_25_2001@hotmail.com National Organization for Drug Control and Research, Cairo, Egypt

2016 Taylor & Francis


2 M. H. SHUKR

amphiphiles have been used in niosomal formulations to improve dried thin lipid film was then hydrated with 10 ml of phosphate
their properties such as stability and drug-loading capacity. buffered saline (PBS, pH 7.4), by rotating the flask in a water bath
Mixed niosomes were developed using pluronics F127 (EO100 using a rotavapour under normal pressure in order to ensure
PO65EO100) and L64 (EO13PO30EO13) with span 40 and span 60. complete hydration of the film. The niosomal suspension was left to
Pluronics are widely used triblock copolymers consisting of hydro- mature overnight at 4  C (Bayindir and Yuksel, 2010).
philic poly (ethylene oxide) (PEO) and hydrophobic poly (propylene
oxide) (PPO) groups. These polymers were chosen due to their
ability to interact with the body membranes and hydrophobic Determination of drug encapsulation efficiency (% EE)
surfaces, thus enhancing the drug transport across cellular barriers The amount of drug entrapped into niosomal vesicles was
(Tavano et al., 2013). The addition of cholesterol into the niosomal determined by ultracentrifugation, 1 mL of the niosomal suspension
formulation results in increasing hydrophobicity (Bernsdorff et al., was placed into centrifuge tubes and ultracentrifuged for 1 h at
1997) and stability (Gregoriadis and Davis, 1979) of the bilayer but 15 000rpm at 4  C (Ultra centrifuge 5417R, Eppendorf, Hamburg,
decreasing membrane permeability (Kirby et al., 1980) which Germany), in order to separate unentraped drug. The niosomes
ultimately leads to improvement in encapsulating the lipophilic were separated from the supernatant and 0.1 ml of the supernatant
drug into bilayers as vesicles assembled. Higher entrapment was added to 10 ml methanol in a 10 ml volumetric flask and the
efficiency was achieved at 1:1 cholesterol/surfactant molar ratio concentration of VCZ was determined spectrophotometrically at
compared with the higher cholesterol molar ratio or niosomal 256 nm. The % EE was calculated according to the following
formulations without any added cholesterol (Balakrishnan et al., equation (Aggarwal and Kaur, 2005):
2009). In this study, the ratio of surfactant and cholesterol was
adjusted to 1:1. % EE Ct  Cf =Ct   100
Downloaded by [Gazi University] at 05:48 24 January 2016

Materials and methods where Ct is the concentration of total VCZ and Cf is the concen-
Materials tration of free VCZ.

VCZ (99%) was kindly provided by Pfizer. Diazepam (as an internal Vesicle size, size distribution and zeta potential
standard) was kindly provided by Amoun Pharmaceutical Industries
Co., Cairo, Egypt. Span 40, span 60 and cholesterol (CH) were The niosomes size and polydispersity index (PI) were determined
purchased from Sigma Chemical Co. (St. Louis, MO). Pluronic L64, using Master sizer (Malvern Instruments, Malvern, Worcestershire,
pluronic F127 and sodium alginate (ALG) were purchased from UK). The diluted niosomal suspension (1 ml) was vortex mixed for
Sigma Aldrich (St. Louis, MO). Sodium carboxy methylcellulose (CMC 5 min then placed in the cell of the master sizer and the size and
Na) medium viscosity: Biochemica, Baden-Dattwil, Switzerland. polydispersity index (PI) were determined. The zeta potential was
Spectra Por semi-permeable membrane tubing (molecular measured with photon correlation spectroscopy (PCS) using
weight cut off of 12 00014 000) was purchased from Spectrum Malvern Zetasizer (Malvern, UK). Each sample was measured in
Medical Industries (Los Angeles, CA). Chloroform, sodium bicarbon- triplicate.
ate, calcium chloride, potassium chloride and sodium chloride were
from El-Nasr Pharmaceutical Chemicals Co., Cairo, Egypt. Acetonitrile Transmission electron microscopy (TEM)
and methanol (HPLC grade) were purchased from Sigma Aldrich
Chemical Co. (St. Louis, MO). All other chemicals were of analytical The morphological features of the prepared niosomes were examined
grade and used as received. via TEM. One drop of each drug-loaded niosomal formulation was
loaded onto a copper grid and the excess was removed with a filter
paper. The formulation was negatively stained with one drop of
Preparation of niosomes
phosphotungstic acid aqueous solution (2%, w/v) for 3 min.
Drug-loaded niosomes were prepared using film hydration tech- Examination of the grid was achieved via a transmission electron
nique. In a flask of a rotary evaporator (Buchi R-110 Rotavapor, microscope (TEM; Tecnai G20, FEI, Hilsboro, OR, Super twin, double tilt).
Flawil, Switzerland), the mixture of surfactants (pluronic F127,
pluronic L64, span 40 and span 60), drug and cholesterol were In vitro release
dissolved in chloroform at the molarities shown in Table 1. Then the
previous solution was completely dried in a rotary evaporator at a The in vitro drug release studies of the prepared niosomes were
rotating speed of 70 rpm and water bath maintained at 50  C. The performed, in triplicate, using a USP dissolution tester (Apparatus I,
Hanson SR6, Chatsworth, CA), at 34 0.5  C to simulate the ocular
Table 1. Composition of the prepared niosomes. surface temperature (Miyazaki et al., 2001). One millilitre of each
formula was placed in a glass cylindrical tube (2 cm internal
Formula Span Span Pluronic Pluronic
Code 60 40 L64 F127 Cholesterol VCZ diameter and 5 cm length) tightly covered with a pre-soaked
Spectra Por semi-permeable membrane tubing from one end. The
F1 mM 10 10 10
mg 44.17 39.57 35.7 loaded tubes were attached to the shafts of the USP Dissolution
F2 mM 5 5 10 10 tester apparatus from the other end (Abd-Elbary et al., 2008). The
mg 22.08 643.32 39.57 35.7 shafts rotated at a speed of 25 rpm in simulated tear fluid (pH 7.4,
F3 mM 5 5 10 10 100 ml). The simulated tear fluid (based on the electrolyte compos-
mg 22.08 147.83 39.57 35.7
ition of tear fluid) was prepared using 6.8 g of NaCl, 2.2 g of NaHCO3,
F4 mM 10 10 10
mg 41.14 39.57 35.7 0.084 g of CaCl22H2O and 1.4 g of KCl in 1 L of ultrapure water.
F5 mM 5 5 10 10 These amounts are equal to 142 mM of Na+, 19 mM of K+ and 0.6 mM
mg 20.57 643.32 39.57 35.7 of Ca2+ with an osmolality of 288 5 mmol/kg (Paulsson et al., 1999).
F6 mM 5 5 10 10 Periodically, 1 ml samples were withdrawn and replaced with fresh
mg 20.57 147.83 39.57 35.7
medium at predetermined time intervals of 0.25, 0.5, 1, 1.5, 2, 3, 4, 5,
JOURNAL OF MICROENCAPSULATION 3

6, 7 and finally at 8 h. The drug-released percentages were and coated with goldpalladium under an argon atmosphere using
determined spectrophotometrically at 256 nm. a gold sputter module in a high-vacuum evaporator and then the
surface was examined. The morphological features of the hydrated
inserts were examined via TEM.
Ocular insert preparation
Polymer solutions were prepared by dissolving specified amounts of
In vitro drug release
ALG and CMC Na in distilled water to produce final concentration of
2% w/v. The selected drug-loaded niosomeal formulation (drug The in vitro release of VCZ from different ocular inserts was carried
equivalent to 0.25%w/v) was added. The formed polymeric solutions out in triplicate, using a USP dissolution tester (Apparatus I, Hanson
were sonicated to remove air bubbles. Aliquots of 0.4 ml were SR6, Chatsworth, CA), at 34 0.5  C to simulate the ocular surface
poured into each of the pockets of a PVC blister with a diameter of temperature (Miyazaki et al., 2001). One insert was placed in a glass
10 mm and frozen at 25  C for 24 h. The frozen inserts were placed cylindrical tube (2 cm internal diameter and 5 cm length) tightly
then in a lyophiliser for 24 h using a Novalyphe-NL 500 Freeze Dryer covered with a pre-soaked Spectra Por semi-permeable mem-
(Savant Instruments, Holbrook, NY) with a condenser temperature brane tubing from one end. The procedure was continued as
of  45  C and a pressure of 7  10  2 mbar. The lyophilised inserts previously mentioned.
were stored in desiccators over calcium chloride (0% relative
humidity) at room temperature until further use (Bertram and
Bodmeier, 2012). Statistical analysis
A one-way analysis of variance (ANOVA) followed by Tukeys
pairwise test at 5% significance level was used to test for statistical
Characterisation of the inserts loaded with VCZ niosomes
Downloaded by [Gazi University] at 05:48 24 January 2016

significance between the prepared formulations using GraphPad


Water uptake and mass loss of the inserts Software Version 3.05 (GraphPad Software Inc., San Diego, CA).
A sponge with dimensions of 5 cm  6.5 cm  3 cm was fully soaked
in the hydration medium STF and placed in a petri-dish filled with In vivo evaluation of VCZ-loaded in situ gelling ocular inserts
the same medium to a height of 1 cm to keep the sponge soaked
during the experiment. Round filter paper (d 55 mm, Schleicher The potential ocular irritancy and/or damaging effects of the
Schuell GmbH, Dassel, Germany) was also soaked in medium and selected insert were evaluated by observing them for any redness,
positioned on the top of the sponge. The experimental set up was inflammation or increased tear production, upon application to the
equilibrated for 30 min. Accurately weighed inserts were placed on eyes of albino rabbits. The selected inserts were pre-sterilised prior
the filter paper and the water uptake was determined as weight to application by irradiation with a short-wavelength UV beam
increase of the insert (weight of hydrated insert and wet filter paper (254 nm) on both sides for 10 min using a hand-held UV lamp
minus weight of wet filter paper) over time normalised to the initial (UVGL-58 UVP, Upland, Cambridge, UK). Insert was inserted into
dry insert weight (Bertram and Bodmeier, 2006). the conjunctival sac in the right eye of three rabbits and the left
eye was left without any treatment as a control. Both eyes of the
rabbits under test were examined for any irritation signs, such as
Surface pH conjunctival corneal oedema and/or hyperaemia on the basis of
direct visual observation using a slit lamp, before treatment, and 1,
The inserts were left to swell for 2 h on the surface of agar plate.
24 and 48 h after insertion (Tamilvanana and Benitab, 2004; Colo
Agar solution was prepared by dissolving 2%w/v agar in STF by
et al., 2001).
heating under stirring, then poured into a petri dish for gel
formation at room temperature. Surface pH was measured by means
of a pH paper placed on the surface of the swollen inserts (Nafee Estimation of VCZ in the rabbit aqueous humour
et al., 2003). The measurements were performed in triplicate.
The in vivo study was carried out to compare the pharmacokinetics
of VCZ in the rabbit aqueous humour following ocular instillation of
Insert mucoadhesion properties a drug suspension and selected insert. The experimental design was
in agreement with the ethical principles of EU Directive 2010/63/EU
Mucoadhesion was performed by adapting the displacement
for the use of animals in scientific researches. Ten New Zealand
method of Bertram and Bodmeier (2006). Agar/mucin solution
albino rabbits weighing 22.5 kg were used in this study and were
(1%w/w, and 2% w/w, respectively) in STF was casted on a petri
randomly divided into two groups. The lower conjunctival sac of the
plate and left to gel at 48  C in a refrigerator for 3 h. The inserts
right eye of each rabbit in the first group was treated with the drug
were placed on the top of the agar/mucin gel, casted on a glass
suspension (1%, w/v, 50 ml) in two instillations at 5 min intervals
plate in an incubator at 37  C. The inserts were moved downward
using a micropipette without touching the eyes or irritating the
due to the gravity after the glass plate was turned into a vertical
corneal surface. While those of the other group were treated with
position. The displacement in centimetre was measured after 2 h
the selected formulation containing the same amount of VCZ in the
(n 3). The adhesive potential was inversely related to the in situ gelling insert. The rabbits were systemically anaesthetised
displacement of the insert. with intramuscular injections of ketamine hydrochloride (50 mg/kg)
in combination with a relaxing agent xylazine (10 mg/kg) (El-Laithy
Scanning electron microscopy (SEM) et al., 2011). They were locally anaesthetised with benoxinate HCl
(0.4%, w/v, 2 drops) at 0.5, 1, 2, 4, 8 and 10 h post-instillation,
The shape and the surface morphology of selected ocular insert aqueous humour samples (0.15 ml) were withdrawn using an insulin
were performed by SEM (JXA-840 A, Tokyo, Japan). Inserts were cut syringe (1.0 ml) fitted with a 29 G needle. The samples were
with a razor blade to expose the inner structure, fixed on supports collected and stored at  20  C until analysed.
4 M. H. SHUKR

Sample preparation vesicles decreased to 122.7 and 157.4 nm, respectively, with a
polydespersity index of 0.28. However, the zeta potential was
The thawed aqueous humour samples were spiked with Diazepam, 29.8 mV and 38.5 mV.
mixed (0.10 ml) with acetonitrile (0.20 ml) and vortexed for 30 s. The results (Table 2) revealed that the mean vesicles size of
After centrifugation for 15 min at 2000  g, the organic phase in the niosomes significantly decreased (p50.05) in the order of span
upper layer was collected. A portion of 20 ml was used for HPLC. 604span 404span 60 with pluronic L644span 60 with pluronic F
1274span 40 with pluronic L 644span 40 with pluronic F 127.
HPLC analysis of VCZ in rabbit aqueous humour
A previously reported HPLC method for the determination of VCZ in Entrapment efficiency (EE%)
human plasma was adopted for the determination of the drug Table 2 shows the results of the percent entrapment efficiency. The
pharmacokinetics in the aqueous humour of the rabbits eyes
entrapment efficiency followed the trend Sp 60 with pluronic
(Khoschsorur et al., 2005). A Hypersil C18 column (4.6 mm  250 mm,
L644Sp 604Sp 404Sp 60 with pluronic F1274Sp 40 with pluronic
5 mm, Thermo Scientific, Uppsala, Sweden) was used as the analytical
L644Sp 40 with pluronic F127. From the results tabulated, it is clear
column maintained at 40  C. The mobile phase consisted of 50 mM
that niosomes prepared with span 60 alone exhibited higher EE
phosphate buffer, pH 6.0 (adjusted with 1 M KOH), acetonitrile, and
(64.6%) than niosomes prepared with span 40 alone (56.5%). The
methanol (35:45:20) delivered at 1.7 mL/min. UV-detection was
highest EE (75.6%) was exhibited with mixed niosomes prepared
performed at 255 nm and Diazepam was employed as the internal
with span 60 and pluronic L64.
standard (1 mg/ml), and the injected volume was 30 ml.
Downloaded by [Gazi University] at 05:48 24 January 2016

TEM
Pharmacokinetic analysis
Figure 1(a) and (b) shows that the niosomes were homogenous and
The non-compartment model is used to evaluate pharmacokinetic
had a spherical shape with a smooth surface.
parameters of VCZ using WinNonlin1 5.3 software (Pharsight
Corporation, Mountain View, CA). The maximum VCZ concentration
in the aqueous humour of the rabbits eyes (Cmax, mg/ml) and the
In vitro release
time to reach Cmax (Tmax, h) were calculated from the individual
aqueous humour concentrationtime curves. The trapezoidal rule In vitro release profiles of VCZ from different formulations are
method was adopted to calculate the area under curve from zero to illustrated in Figure 2. F4 formulation prepared with span 40 alone
10 h (AUC010, mg h/ml). The increase in the ocular bioavailability exhibited the highest percentage released (85%) after 480 min,
(folds) was calculated from the AUC010 of the S2 insert and that of followed by F1 prepared with span 60 alone (80%), then mixed
the drug suspension. niosomes prepared with span 40 and span 60 with pluronic F127
(77% and 75%, respectively), and finally mixed niosomes prepared
with span 40 and span 60 with pluronic L64 (74% and 63%,
Results respectively). From the previous results, F3 mixed niosomes
Optimisation of niosomal formulation prepared with span 60 and pluronic L64 was selected for the
preparation of ocular inserts.
Vesicle size, polydispersibility index (PI) and zeta potential
The average vesicle size and zeta potential of niosomes are shown in
Table 2. Formulations F1 and F4 (prepared with span 60 and span 40 Characterisation of the inserts loaded with VCZ niosomes
alone) showed the largest vesicle size (478 nm and 415 nm, Figure 3 shows the sponge-like structure of the ocular inserts.
respectively) with polydispersity indexes of 0.36 and 0.67, with a Surface pH measurements were in the range of 7 0.2 (Table 3). TEM
comparable zeta potential of 36.5 mV and 35.5 mV, respectively. was used to investigate the morphology of the niosomes within the
When a mixture of span 60 and pluronic L64 was used (F3), it was inserts after hydration. Figure 4(a) shows that no change was
observed that both the size of the vesicles and PI were decreased to observed in the shape of the niosomes. However, the vesicle size of
214 nm and 0.28, respectively. However, the zeta potential was
the niosomes (Figure 4b) was significantly increased (p50.05) from
increased to 38.5 mV. F2 formulation (prepared with span 60 and
niosomal suspension to niosomes within insert after hydration.
pluronic F 127) showed the smallest zeta potential (22.4 mV) and
Figure 5 shows the results of water uptake studies, over the study
largest PI (0.68) with an average size of 127.8 nm.
period (5 h), the two ocular inserts hydrated without dissolution and
On the other hand, for the mixture of span 40 with pluronic F127
CMC Na insert exhibited higher moisture uptake than AlG insert.
(F5) and pluronic L64 (F6), it was observed that the size of the
CMC Na and ALG inserts showed high water uptake.
The vertical displacement of inserts on an agar/mucin gel was
Table 2. Entrapment efficiency, mean vesicle size, polydispersibility index (PI) and
zeta potential of different niosomes (results are expressed as mean values SD, used as a measure of bioadhesion potential. ALG shows significantly
n 3). (p50.05) stronger adhesive forces (0.4 cm) compared with CMC
Formula Vesicle size (0.7 cm). The release profiles of VCZ in STF pH 7.4 from the two in
Code (nm) Zeta Potential EE% PI situ gelling ocular insert are shown in Figure 6. The two formulation
F1 478.0 2.3  36.5 0.2 66.6 1.5 0.36 0.01 exhibited prolonged drug release for at least 8 h.
F2 127.8 1.8  22.4 0.13 58 1.7 0.68 0.012
F3 214 1.9 38.5 0.25 75.6 1.3 0.28 0.012
F4 415 3.2 35.5 0.23 61.5 1.7 0.67 0.023 In vivo evaluation of VCZ-loaded in situ gelling ocular inserts
F5 122.70 2.1 29.9 0.19 53 1.6 0.28 0.011
F6 157.40 1.83 35.7 0.23 49.7 1.8 0.28 0.01 It was observed that over the study period (48 h), the selected
formulation (S2) produced non-irritant effect on the rabbits eyes, it
JOURNAL OF MICROENCAPSULATION 5

Figure 1. Transmission electron micrographs of F3 niosomal formulation.


Downloaded by [Gazi University] at 05:48 24 January 2016

Figure 2. In vitro drug release from niosomal formulation (a) prepared with span 60 and (b) with span 40 in simulated tear fluid (pH 7.4) at 34 0.5  C (mean SD, n 3).

Figure 3. Scanning electron micrographs of the ocular insert prepared from Na CMC (S1) and ALG (S2).

can be accomplished that the tested formulation was well tolerated Table 3. Concentration of the corresponding polymer solutions, pH and
mucoadhesion of the prepared ocular inserts.
showing no evidence of redness, inflammation, or increased tear
production effects to the ocular surface. Insert Polymer concentration pH mean SD Displacing (cm)
The drug pharmacokinetics in the aqueous humour follow- code (% w/w) (n 3) mean SD (n 3)
ing ocular insertion of S2 in situ gelling ocular insert and S1 2% Na CMC 6.8 0.31 0.7 0.1
the drug suspension was evaluated in rabbits (Figure 7). S2 2% Na alginate 6.9 0.25 0.4 0.1
6 M. H. SHUKR

Figure 4. Transmission electron micrographs of S2 in situ gelling ocular insert after hydration.
Downloaded by [Gazi University] at 05:48 24 January 2016

Figure 7. Aqueous humour concentrationtime profiles of VCZ following insertion


of S2 in situ gelling ocular insert and instillation of the drug suspension in rabbits
(mean SD, n 5, p50.01).
Figure 5. Water uptake profiles of the in situ gelling VCZ ocular inserts.

Discussion
Preparation of niosomes requires a deep understanding of the
characteristics of the principal ingredients and their effects on the
physicochemical properties and stability of the resultant niosomes.
A variety of non-ionic surfactants and their combinations have been
reported to have great potential to accommodate many drugs in
niosomes (Giddi et al., 2007).
The results are in good agreement with the literature (Kaur et al.,
2008), regarding stability of small niosomes with optimum zeta
potential560 mV. It also indicates the advantage of using pluronic
F127 and pluronic L 64/span 60 and span 40 mixture (1:1) in
producing smaller and physically stable niosomes. All the formula-
tions showed negative zeta potential values, which could reflect
adsorption of counter ions or preferential adsorption of hydroxyl
ions at the niosomal surface (Shahiwala and Misra, 2002). The
Figure 6. In vitro drug release from in situ gelling ocular inserts in simulated tear
fluid (pH 7.4) at 34 0.5  C (mean SD, n 3). highest zeta potential was observed for the niosomes prepared with
span 60 and pluronic L 64, indicating that the mixed niosomes (F3)
were more stable against aggregation and fusion.
The hydrophilelipophile balance (HLB) values of non-ionic
The differences between the two treatments were mathematically surfactants can be a representative of the vesicle forming ability.
evaluated by the calculation of Cmax, Tmax and AUC(010). The Cmax Surfactants of HLB values between 4 and 8 (e.g. span surfactants)
and Tmax values representing the drug suspension and the in situ were reported to be compatible with vesicle formation (Uchegbu and
gelling ocular insert were 1170.26 ng/ml after 2 h and 2203 ng/ml Vyas, 1998). Addition of applied cholesterol completes the lipophilic
after 1 h, respectively. The differences between the two treatments moiety of non-ionic surfactants with high HLB values to form vesicles
were statistically significant (p50.01). These differences were (Masotti et al., 2010). For surfactants with higher HLB values, the
proved to be statistically significant, p50.01. The increase in the minimum amount of cholesterol necessary to form vesicles increases
ocular bioavailability of VCZ, judged from the AUC(010), was 5.91- in order to compensate the larger hydrophilic head group which
fold. increases the lipophilic behaviour of the lipid bilayer (Kumarn and
JOURNAL OF MICROENCAPSULATION 7

Rajeshwarrao, 2011). This may explain the observed smaller particle The values of surface pH indicating that the prepared inserts is
size for niosomes prepared with span 60 or span 40 and pluronic compatible with the physiological pH of the surface of the eye, this
F127 or pluronic L64 than those prepared with span 60 and span 40 minimises any possible ocular surface irritation, tearing and reflex
alone. The HLB values were 6.7, 4.7, 22 and 8 for span 40, span 60, blinking.
pluronic F127 and pluronic L64, respectively. Increasing the vesicle size of niosomes might be attributed to a
The decrease in the vesicle size that was observed with mixed fusion of vesicles. The swelling behaviour of in situ gelling insert
niosomes prepared with span 40 and span 60 with pluronics (L64 governs its bioadhesion and drug release pattern. The faster the
and F 127) could be explained as the concentration of added swelling of the polymer, the faster the initiation of diffusion and
cholesterol was more efficient with niosomes prepared with span 60 formation of adhesive bonds resulting in faster initiation of
and span 40 alone than with mixed niosomes and smaller vesicle bioadhesion (Singh et al., 2010). The uptake of water by in situ
sizes were obtained. As the concentration of cholesterol increases, gelling inserts is a crucial step for the transformation into the gel
the vesicle size also increases; this may be attributed to the fact that and for adhesion to the mucosa.
cholesterol increases the width of vesicular bilayers and conse- The high water uptake observed for CMC Na and ALG inserts
quently increases the vesicle size (Hosny, 2010). could be greatly attributed to the ionic nature of the polymers,
EE is one of the important parameters in the design of vesicular allowing high tendency towards water uptake.
formulations. The higher EE exhibited with span 60 may be CMC Na has an abundance of hydroxyl and ether groups along
explained due to the effect of HLB of the surfactant, the lower the their length, which are responsible for the higher water uptake
HLB the higher will be the drug EE and stability. Because higher HLB properties (Mansour et al., 2008). When CMC Na is exposed to an
values reflect a low hydrocarbon chain volume in comparison with a aqueous medium, it undergoes rapid hydration and chain relaxation
hydrophilic surface area, they result in niosomes that are difficult to to form a viscous gelatinous layer (gel layer).
form (Parthasarathi et al., 1994). Cholesterol is commonly used as an For a polymer to show good adhesive characteristics to the
Downloaded by [Gazi University] at 05:48 24 January 2016

additive in niosomal systems, to influence the stability and mucoploysaccharide components of the mucin produced by the
permeability of the membrane, which is mainly due to interaction conjunctival goblet cells, the polymer must demonstrate one or
between surfactant and cholesterol, through hydrogen bonding more of the following characteristics: hydrogen-binding capacity,
between its hydroxyl group and surfactants hydrocarbon chain anionic charge, high molecular weight, sufficient chain flexibility and
which increase the mechanical stiffness of the membranes and spreading ability on mucus coat (Gilhotra et al., 2009).
membrane cohesion (Aripin et al., 2012). Wetting has been reported to be essential for establishment of
The highest EE observed with F3 mixed niosomes (span 60 and intimate contact between the mucoadhesive and mucin/tissue to
pluronic L64) may be due to hydrogen-bonding association develop strong adhesive bond (Wang and Bazos, 1983) and
between the straight-chain structures of pluronic (L64) and span prolonged ocular drug delivery. Both ALG and CMC have free
60 (C16) during the formation of noisome vesicles. This interaction hydroxyl groups available for hydrogen binding with the conjunc-
seemed to occur at lower extent between span 60 with pluronic tival mucin coat.
F127 (it has a larger molecular weight of 11 500) and span 40 with Furthermore, the stronger adhesive forces for ALG could be
and pluronics. ascribed to the conformational arrangements of D-mannuronic acid
TEM was used to investigate the morphology of the niosomes. (M blocks) and L-guluronic acid (G blocks) of ALG chains that are
The images also confirmed that the niosomes were of nanometre favourable for interaction with the mucin coat, as well as the higher
size. density of carboxylate groups and higher molecular weight
The results of drug release were in agreement with the results of (100 kDa) for ALG, compared with CMC (90 kDa) (Skjak-Brak, 1992).
EE%, F3-mixed niosomes prepared with span 60 and pluronic L64 The polymeric materials become adhesive with hydration while
exhibited the highest EE % and showed the lowest percent of drug excessive moisture uptake leads to reduced mucoadhesiveness
release. This can be explained as previously mentioned due to because water molecules bind to polymer groups required for
hydrogen-bonding association between the straight-chain struc- adhesion (Deasy and ONeill, 1989).
tures of pluronic L64 and span 60 (C16) during the formation of ALG (S2) absorbs a significant amount of STF to hydrate, swells
noisome vesicles. and forms a stable hydrogel upon exposure to the divalent cations
It was apparent that the incorporation of VCZ in mixed niosomes Ca + 2 present in STF. The drug which is embedded in the ALG insert
led to significant slower release profiles (p50.05) compared with is now immobilised in the polymer, therefore, the in situ gel-forming
niosomes prepared with span 60 and span 40 alone (F1 and F4, insert acts as a reservoir for the drug (Mishra and Gilhotra, 2008).
respectively). The acceleration of the early portion of the release curve Drug release from CMC Na ocular insert (S1) was relatively slower
is the result of VCZ desorption from the surface of niosomes while the than from ALG ocular insert (S2) (Figure 6).
drug release in the slower phase was regulated by diffusion through This may be explained by referring to the water uptake data,
the swollen niosomal bilayers (Pardakhty et al., 2007). where CMC Na exhibited higher water uptake (Figure 5). Although
F3 mixed niosomes were selected due to their highest encap- the marked increase in surface area during swelling can promote
sulation efficiency; optimum particle size with smallest variation in drug release, the increase in diffusional path length of the drug may
size, which indicated by smallest PI and suitable prolonged release paradoxically delay such release. Further, the thick gel layer formed
pattern of the drug. on the swollen insert surface is capable of preventing matrix
Ocular inserts are prepared by freeze-drying of aqueous solutions disintegration and controlling additional water penetration
containing polymeric carriers and drug. This production procedure (Rodriguez et al., 2000).
ensures the formation of highly porous polymeric sponges into According to the above results in situ ocular insert S2 was chosen
which VCZ niosomes are embedded. depending on their superior mucoadhesive properties, appropriate
The sponge-like structure of the ocular inserts is an important water uptake and suitable prolonged release profile and hence was
parameter to ensure rapid hydration and gelation of the insert at the subjected to further in vivo evaluation.
ocular mucosa resulting in a larger surface/contact area and The aqueous humour concentrationtime profiles of VCZ clearly
increased water uptake by capillary forces and to a reduced foreign indicate the ability of the in situ gel to control the drug release rate
body sensation when compared with other solid dosage forms. upon insertion. The delay in Tmax (from 1 to 2 h) for the drug
8 M. H. SHUKR

suspension and the in situ gelling inserts, respectively, could Balakrishnan P, Shanmugam S, Lee WS, Lee WM, Kim JO, Oh DH,
indicate the controlled release characteristics of the latter. The Kim DD, Kim JS, Yoo BK, Choi HG, et al. Formulation and in vitro
higher ocular bioavailability of the in situ gelling ocular insert could assessment of minoxidil niosomes for enhanced skin delivery. Int J
be related to its ability to prolong the residence time on the corneal Pharm, 2009;377(1-2):18.
epithelial layer. This would enable better transcorneal drug pene- Basaran E, Demirel M, Sirmagul B, Yazan Y. Cyclosporine-A
tration to the ocular internal tissues. incorporated cationic solid lipid nanoparticles for ocular delivery.
J Microencapsul, 2010;27:3747.
Bayindir ZS, Yuksel N. Characterization of niosomes prepared with
Conclusion various nonionic surfactants for paclitaxel oral delivery. J Pharm
A novel in situ gelling ocular inserts loaded with VCZ niosomes was Sci, 2010;99:204960.
developed and characterised in this study. Niosomes made up of Bernsdorff C, Wolf A, Winter R, Gratton E. Effect of hydrostatic
mixed surfactants (prepared with span 60 and pluronic L64: choles- pressure on water penetration and rotational dynamics in
terol at a molar ratio of 1:1) proved to be effective carriers and phospholipidcholesterol bilayers. Biophys J, 1997;72(3):126477.
exhibited high EE and relatively small vesicle size and prolonged drug Bertram U, Bernard MC, Haensler J, Maincent P, Bodmeier R. In situ
release. The niosomal suspension was further developed to form in gelling nasal inserts for influenza vaccine delivery. Drug Dev Ind
situ gelling ocular insert by the freeze-drying method. The morph- Pharm, 2010;36:58193.
ology, surface pH, mucoadhesion and release study were evaluated Bertram U, Bodmeier R. Effect of polymer molecular weight and of
for the VCZ-loaded niosomal ocular insert. ALG in situ gelling insert polymer blends on the properties of rapidly gelling nasal inserts.
(S2) showed prolonged drug release with superior mucoadhesion and Drug Dev Ind Pharm, 2012;38(6):65969.
Bertram U, Bodmeier R. In situ gelling, bioadhesive nasal inserts for
appropriate water uptake, and it was selected for further in vivo
extended drug delivery: In vitro characterization of a new nasal
Downloaded by [Gazi University] at 05:48 24 January 2016

evaluation. The sterilised S2 in situ gelling ocular insert was non-


dosage form. Eur J Pharm Sci, 2006;27:6271.
irritant to rabbits eyes, succeeded in controlling the rate of VCZ
Bhatta RS, Chandasana H, Chhonker YS, Rathi C, Kumar D, Mitra K,
release in the rabbit aqueous humour and improved the ocular drug
Shukla PK. Mucoadhesive nanoparticles for prolonged ocular
bioavailability, relative to the drug suspension.
delivery of natamycin: In vitro and pharmacokinetics studies. Int J
The VCZ in situ gelling niosomal inserts could prove to be a good
Pharm, 2012;432:10512.
alternative to conventional niosomes formulations, combine the
Colo DG, Burgalassi S, Chetoni P, Fiaschi MP, Zambito Y,
advantages of solid, single unit dosage forms, such as high stability
Saettone MF. Gel-forming erodible inserts for ocular controlled
and dosing accuracy, with those of gel preparations, which avoid
delivery of ofloxacin. Int J Pharm, 2001;215:10111.
foreign body sensation and allow prolonged drug delivery.
Dai YK, Zhou R, Liu L, Lu Y, Qi JP, Wu W. Liposomes containing bile
salts as novel ocular delivery systems for tacrolimus (FK506):
Declaration of interest In vitro characterization and improved corneal permeation. Int J
Nanomed, 2013;8:192133.
The author reports no conflicts of interest. The author alone is Deasy PBO, Neill CT. Bioadhesive dosage form for peroral admin-
responsible for the content and writing of the article. istration of timolol base. Pharm Acta Helv, 1989;64:231.
deSa FA, Taveira SF, Gelfuso GM, Lima Em Gratieri T. Liposomal
voriconazole (VOR) formulation for improved ocular delivery.
References
Colloids Surf B Biointerfaces, 2015;133:3318.
Abdelbary G, El-gendy N. Niosome-encapsulated gentamicin for oph- El-Laithy HM, Nesseem DI, El-Adly AA, Shoukry M. Moxifloxacin-
thalmic controlled delivery. AAPS PharmSciTech, 2008;9:7407. Gelrite in situ ophthalmic gelling system against photodynamic
Abdelbary A, El-laithy HM, Tadros MI. Sucrose stearate-based therapy for treatment of bacterial corneal inflammation. Arch
proniosome-derived niosomes for the nebulisable delivery of Pharm Res, 2011;34:166378.
cromolyn sodium. Int J Pharm, 2008;357:18998. Farid RM, Etman MA, Nada AH, Ebian AA. Formulation and in vitro
Abdelkader H, Wu Z, Al-Kassas R, Brown JE, Alany RG. Preformulation evaluation of salbutamol sulphate in situ gelling nasal inserts.
characteristics of the opioid growth factor antagonist-naltrexone AAPS Pharm SciTech, 2013;14:71218.
hydrochloride: Stability and lipophilicity studies. J Drug Del Sci Giddi H, Arunagirinathan M, Bellare J. Self-assembled surfactant
Technol, 2011a;21:15763. nano-structures important in drug delivery: A review. Indian J Exp
Abdelkader H, Patel DV, Mc Ghee CN, Alany RG. New therapeutic Biol, 2007;45(2):13359.
approaches in the treatment of diabetic keratopathy: A review. Gilhotra RM, Gilhotra N, Mishra DN. Piroxicam bioadhesive ocular
Clin Exp Ophthalmol, 2011b;39:25970. inserts: Physicochemical characterization and evaluation in pros-
Abdelkader H, Alani AW, Alany RG. Recent advances in nonionic taglandin-induced inflammation. Curr Eye Res, 2009;34:106573.
surfactant vesicles (niosomes): Self-assembly, fabrication, charac- Gratieri T, Gelfuso GM, de Freitas O, Rocha EM, Lopez RF. Enhancing
terization, drug delivery applications and limitations. Drug Deliv, and sustaining the topical ocular delivery of fluconazole using
2013;21:87. chitosan solution and poloxamer/chitosan in situ forming gel. Eur
Aggarwal D, Kaur IP. Improved pharmacodynamics of timolol J Pharm Biopharm, 2011;79:3207.
maleate from a mucoadhesive niosomal ophthalmic drug delivery Gregoriadis G, Davis C. Stability of liposomes in vivo and in vitro is
system. Int J Pharm, 2005;290:1559. promoted by their cholesterol content and the presence of blood
Ahmed IS, Aboul-Einien MH. In vitro and in vivo evaluation of a fast- cells. Biochem Biophys Res Commun, 1979;89(4):128793.
disintegrating lyophilized dry emulsion tablet containing griseo- Gupta S, Shrivastava RM, Tandon R, Gogia V, Agarwal PD, Satpathy
fulvin. Eur J Pharm Sci, 2007;32:5868. G. Role of voriconazole in combined acanthamoeba and fungal
Aripin NFK, Park JW, Park HJ. Preparation of vesicle drug carrier from corneal ulcer. Cont Lens Anterior Eye, 2011;34:2879.
palm oil-and palm kernel oil-based glycosides. Colloids Surf B, Hosny KM. Ciprofloxacin as ocular liposomal hydrogel.
Biointerfaces, 2012;95:14453. AAPSPharmSciTech, 2010;11:2416.
JOURNAL OF MICROENCAPSULATION 9

Kapoor Y, Chauhan A. Ophthalmic delivery of Cyclosporine A from delivery of pilocarpine hydrochloride. Int J Pharm, 2001;229:
Brij-97 microemulsion and surfactant-laden p-HEMA hydrogels. Int 2936.
J Pharm, 2008;361:2229. Nafee NA, Ismail FA, Boraie NA, Mortada LM. Mucoadhesive buccal
Kaur IP, Bhandari R, Bhandari S, Kakkar V. Potential of solid lipid patches of miconazole nitrate: In vitro/in vivo performance and
nanoparticles in brain targeting. J Control Release, 2008;127: effect of ageing. Int J Pharm, 2003;264:114.
97109. Pardakhty A, Varshosaz J, Rouholamini A. In vitro study of
Kaur IP, Kakkar S. Topical delivery of antifungal agents. Expert Opin polyoxyethylene alkyl ether niosomes for delivery of insulin. Int
Drug Deliv, 2010;7:130327. J Pharm, 2007;328(2):13041.
Kaur IP, Rana C, Singh M, Bhushan S, Singh H, Kakkar S. Parthasarathi G, Udupa N, Umadevi P, Pillai GK. Niosome encapsu-
Development and evaluation of novel surfactant-based elastic lated of vincristine sulfate: Improved anticancer activity with
vesicular system for ocular delivery of fluconazole. J Ocul reduced toxicity in mice. J Drug Target, 1994;2:17382.
Pharmacol Ther, 2012;28(5):48496. Paulsson M, Hagerstrom H, Edsman K. Rheological studies of the
Khoschsorur GA, Fruehwirth F, Zelzer S. Isocratic high-performance gelation of deacetylated gellan gum (GelriteR) in physiological
liquid chromatographic method with ultraviolet detection for conditions. Eur J Pharm Sci, 1999;9:99105.
simultaneous determination of levels of voriconazole and itra- Rodriguez CF, Bruneau N, Barra J, Alfonso D, Doelker E. 2000.
conazole and its hydroxy metabolite in human serum. Antimicrob Hydrophilic cellulose derivatives as drug delivery carriers: Influence
Agents Chemother, 2005;49(8):356971. of the substitution type on the properties of compressed matrix
Khutoryanskaya OV, Morrison PW, Seilkhanov SK, Mussin MN, tablets. In: Wise DL, ed. Handbook of pharmaceutical controlled
Ozhmukhametova EK, Rakhypbekov TK, Khutoryanskiy VV. release technology. New York: Marcell Dekker, pp. 130.
Hydrogen-bonded complexes and blends of poly(acrylic acid) Shahiwala A, Misra A. Studies in topical application of niosomally
and methylcellulose: Nanoparticles and mucoadhesive films for entrapped Nimesulide. J Pharm Pharmaceut Sci: A Publ Can Soc
Downloaded by [Gazi University] at 05:48 24 January 2016

ocular delivery of riboflavin. Macromol Biosci, 2014;14:22534. Pharmaceut Sci Soc Can Sci Pharmaceut, 2002;5(3):2205.
Kirby C, Clarke J, Gregoriadis G. Effect of the cholesterol content of Shen J, Deng Y, Jin X, Ping Q, Su Z, Li L. Thiolated nanostructured
small unilamellar liposomes on their stability in vivo and in vitro. lipid carriers as a potential ocular drug delivery system for
Biochem J, 1980;186(2):5918. cyclosporine A: Improving in vivo ocular distribution. Int J Pharm,
Kumarn G, Rajeshwarrao P. Nonionic surfactant vesicular systems 2010;402:24853.
for effective drug delivery An overview. Acta Pharm Sin B, Singh M, Tiwary AK, Kaur G. Investigations on interpolymer
2011;1(4):20819. complexes of cationic guar gum and xanthan gum for formulation
Lat A, Thompson GR. Update on the optimal use of voriconazole for of bioadhesive films. Res Pharm Sci, 2010;5:7987.
invasive fungal infections. Infect Drug Resist, 2011;4:4353. Skjak-Brak G. Alginates: Biosyntheses and some structure-function
Liang QF, Jin XY, Wang XL, Sun XG. Effect of topical application of relationships relevant to biomedical and biotechnological appli-
terbinafine on fungal keratitis. Chin Med J, 2009;122:18848. cations. Biochem Soc Trans, 1992;20:2733.
Luschmann C, Tessmar J, Schoeberl S, Strauss O, Framme C, Tamilvanana S, Benitab S. The potential of lipid emulsion for ocular
Luschmann K, Goepferich A. Developing an in situ nanosuspen- delivery of lipophilic drugs. Eur J Pharm Biopharm, 2004;58:
sion: A novel approach towards the efficient administration of 35768.
poorly soluble drugs at the anterior eye. Eur J Pharm Sci, Tavano L, Vivacqua M, Carito V, Muzzalupo R, Caroleo MC,
2013;50:38592. Nicoletta F. Doxorubicin loaded magneto-niosomes for targeted
Mansour M, Mansour S, Mortada ND, Abd Elhady SS. Ocular drug delivery. Colloids Surf B Biointerfaces, 2013;102:8037.
poloxamer based ciprofloxacin hydrochloride in situ forming Uchegbu IF, Vyas SP. Non-ionic surfactant based vesicles (niosomes)
gels. Drug Dev Ind Pharm, 2008;34:74452. in drug delivery. Int J Pharm, 1998;172:3370.
Masotti A, Vicennati P, Alisi A. NovelTween 20 derivatives enable the Wang PY, Bazos MJ. 1983. Polymer surface interactions in the
formation of efficient pH-sensitive drug delivery vehicles for biological environment. In: Mittal KL, ed. Physicochemical aspects
human hepatoblastoma. Bioorg Med Chem Lett, 2010;20(10): of polymer surfaces. New York: Plenum Press, pp. 14398.
30215. Werner U. 2003. In situ gelling nasal inserts for prolonged drug
Mishra DN, Gilhotra RM. Design and characterization of bioadhesive delivery. Berlin: Fachbereich Biologie, Chemie, Pharmazieder
in-situ gelling ocular inserts of gatifloxacin sesquihydrate. Daru, Freien Universitat Berlin.
2008;16:18. Wiebe V, Karriker M. Therapy of systemic fungal infections:
Miyazaki S, Suzuki S, Kawasaki N, Endo K, Takahashi A, Attwood D. A pharmacologic perspective. Clin Tech Small Anim Pract,
In situ gelling xyloglucan formulations for sustained release ocular 2005;20:2507.

You might also like