Video 11

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Journal of Pharmaceutical Investigation

DOI 10.1007/s40005-014-0121-8

RESEARCH ARTICLE

Effects of surfactant type and cholesterol level on niosomes


physical properties and in vivo ocular performance using timolol
maleate as a model drug
Hamdy Abdelkader Usama Farghaly

Hossam Moharram

Received: 5 January 2014 / Accepted: 18 February 2014


The Korean Society of Pharmaceutical Sciences and Technology 2014

Abstract This study aimed at investigating the roles of vesicles or simply niosomes. Niosomes are considered of
both Span surfactant type (having a gel/liquid transition particular interest as they offer several advantages over lip-
temperature range from\10 to 55 C), and cholesterol level osomes. Niosomes are more stable chemically (Uchegbu and
on in vitro characteristics and in vivo performance of Florence 1995; Kaur et al. 2004). They incur lower produc-
timolol maleate niosomes. Span 20, Span 40 and Span 60 tion cost due to the availability of starting materials (Sahin
niosomes were prepared using the thin film hydration 2006). They are biodegradable and non-immunogenic (Kaur
method. Span:cholesterol levels employed were 7:3 mol/mol et al. 2004). Niosomes do not require expensive handling
(a minimum concentration for stable niosomes) and (storing in a freezer and preparation under nitrogen gas).
1:1 mol/mol (a maximum concentration accommodated by Over the last three decades, niosomes of different
niosomes). Niosomes were characterized for size, percent- compositions used for topical ophthalmic drug delivery
age entrapment efficiency, morphology, in vitro release and have increased/prolonged the therapeutic effect while
intra-ocular pressure (IOP) lowering activity in rabbits. The minimizing toxic effects (Abdelbary and El-gendy 2008;
order of the area under IOP-time curve (AUC) was ranked Abdelkader et al. 2013). Niosomal sizes of [10 lm have
as follows: Span 40 [ Span 20 [ Span 60. The AUC and been reported to be optimum for ocular delivery, in order to
IOPmax values for Span 40:cholesterol (7:3) niosomes were carry a significant amount of payload, resist lacrimal
superior due to having a good compromise between thermo- flushing effect and stay longer on the surface of the eye
responsiveness and efficient capacity for loading a signifi- (Uchegbu and Vyas 1998; Abdelkader and Alany 2012).
cant drug cargo, compared with Span 20 and Span 60. Cholesterol, a bilayer membrane stabilizer, is reported to
increase membrane stability, decrease the fluidity of the
Keywords Niosomes  Intraocular pressure  membrane and alter membrane permeability (Uchegbu and
Timolol maleate  Ocular delivery  Span surfactants Florence 1995; Uchegbu and Vyas 1998). However, the
impact of these effects on the in vivo behavior upon
instillation onto the ocular surface is yet to be resolved.
Introduction Alkyl esters, such as sorbitan fatty acid esters (Span)
have been widely used in ocular delivery (Aggarwal et al.
Vesicles consisting of one or more surfactant bilayers 2004; Abdelbary and El-gendy 2008; Kaur et al. 2008) and
enclosing aqueous spaces are called non-ionic surfactant we have recently reported on their good conjunctival and
corneal tolerability (Abdelkader et al. 2012).
Timolol maleate (TM), a water soluble model drug, was
H. Abdelkader (&)  U. Farghaly
Department of Pharmaceutics, Faculty of Pharmacy, used in this study. TM has been the first topical non-
Minia University, Minia, Egypt selective cardio-selective beta-blocker and has remained
e-mail: hamdy2002m@yahoo.com; h.abdelkader@mu.edu.eg the milestone treatment for glaucoma therapy (Schenker
et al. 1999; Sweetman 2009).
H. Moharram
Department of Ophthalmology, Faculty of Medicine, Niosomes encapsulating TM were prepared using the
Minia University, Minia, Egypt reverse phase evaporation method and coated with 0.5 % w/v

123
H. Abdelkader et al.

chitosan solution. The TM niosomes achieved a signifi- The formed niosomes were stored in a fridge for sub-
cantly (1.7 times) higher TM concentration in the aqueous sequent analyses.
humor than that of the drug solution. This was attributed to
better improvement of corneal penetration and prolonged Entrapment efficiency percentage (EE %)
precorneal residence time compared with the TM aqueous
solution (Kaur et al. 2010). The EE % determination method used relied on exhaustive
However, no plausible explanation has yet to be pro- dialysis of the prepared niosomes and subsequent quanti-
vided on the effects of surfactant types or the cholesterol fication of the released drug molecules. Accordingly, 2 ml
levels on drug uptakes by ocular tissues. Therefore, this of niosome dispersion was transferred into a dialysis bag
report aims at studying the effects of surfactant types and and allowed to exhaustive dialysis in 100 ml of PBS twice
cholesterol levels on in vitro and in vivo performance of for 4 h at 4 C. The dialysate solutions were collected and
ocular niosomes for the anti-glaucomic drug TM. Ocular analyzed for TM content spectrophotometrically at 295 nm
bioavailability could be simply monitored through moni- using a UVVis spectrophotometer (Spectronic, Genesys
toring intra-ocular pressure (IOP) of rabbits, this technique 2PC, USA).
has been employed elsewhere to study the ocular bio- The drug EE % was expressed as a percentage and
availability of different glucocorticoids from nano-sus- calculated using Eq. 1 (Nasr et al. 2008):
pensions (Kassem et al. 2007). Specific objectives will look Ao  A
at the effects of both surfactant types and cholesterol levels EE %  100
Ao
on size, Entrapment efficiency percentage (EE %), mor-
phology and IOP lowering activities for the tested where A is the amount of TM in the dialysate solution and
niosomes. Ao is the initial amount of TM used.

Niosome size measurements


Materials and methods
The size of the prepared niosomes was determined by the
Materials laser diffraction technique using a particle size analyzer
(Quantachrome CLIAS00, France). The samples were
Timolol maleate (TM) was a generous gift from EIPICO properly diluted with PBS and measured at 25 C. Nio-
Pharmaceuticals (Cairo, Egypt). Span 60, Span 40, Span somes size was expressed in terms of volume diameter and
20, cholesterol, and cellulose membrane, molecular weight the measurements were done in triplicate and the average
cut-off 12,00014,000, were purchased from Sigma- values were used.
Aldrich, St. Louis, USA. All other solvents and buffer salts
were of analytical grade and used as received. Transmission electron microscopy (TEM)

Methods Selected niosomal systems prepared, were examined under


TEM. A drop of the niosome sample was transferred into
Thin film hydration method (TFH) the copper mesh grids. After the sample was adsorbed
(about 1520 min), the staining dye (potassium phospho-
Niosomes were prepared by adopting the TFH method tungstate) was dripped onto the film. The staining time was
(Azmin et al. 1985). Briefly, a specified amount about 12 min. After drying the copper mesh grids, the
(900 lmol) of Span 60, Span 40 or Span 20: cholesterol at morphology of the investigated niosomes was visualized
a molar ratio of 1:1 and 7:3 was dissolved in chloroform (Liu and Guo 2007).
and rotary evaporated (Heidolph, Laborota 4000, GmbH,
Germany) to form a thin lipid film. The dried surfactant/ In vitro release studies
lipid film was kept overnight in order to remove any
residual traces of the organic solvent. The lipid film was A sample of 2 ml of the prepared niosomal dispersions was
then hydrated with either 8 ml of phosphate buffer saline placed on a semi-permeable standard cellophane mem-
(PBS) pH 7.4 or TM solution (5 mg/ml) in PBS at 200 rpm brane (previously immersed in PBS for 24 h). The loaded
for 2 h. The selected hydration time at 2 h was based on membrane was stretched over the lower end of an open
preliminary studies. Longer hydration time did not show glass tube of 3 cm diameter and fixed with an elastic band.
further improvement in terms of TM loading. The resultant The glass tube was then immersed in a 50-ml beaker
niosomal dispersion was set aside for at least 2 h at room containing 20 ml PBS (NaCl 137 mM, Na2HPO4 10 mM,
temperature, to allow the vesicles membrane to anneal. KH2PO4 1.47 mM, KCl 2.68 mM) at pH 7.4 in such a

123
Effects of surfactant type and cholesterol level on niosomes

Table 1 Summary of the rabbit groups recruited in the study Statistical analysis
Group Right eye Left eye
To compare mean values of Tmax, IOP and IOPmax for the
Group 1 TM solution (0.25 % w/v) Span 20 7:3 prepared niosomes and the control, unpaired t test and
Group 2 Span 40 7:3 analysis of variance (ANOVA) were carried out at a 5 %
Group 3 Span 40 1:1 significance level using Graph Pad Software, USA) (1995).
Group 4 Span 60 7:3
Group 5 Span 60 1:1
Results and discussion

manner that the membrane was located just below the Span 20, Span 40 and Span 60 surfactants, having melting
surface of the buffer solution. This in-house designed dif- points of 10, 45, and 54 C respectively, were investigated
fusion unit was placed in a thermostatically controlled for their ability to form niosomes in combination with two
shaking water-bath adjusted at 35 0.1 C with a constant different levels of cholesterol at molar ratios of 7:3 and
shaking at 100 rpm to avoid any development of a con- 1:1 mol/mol, forming a cholesterol concentrations in
centration gradient. At appropriate sampling intervals an bilayers of 3050 % mol/mol respectively. In a previous
aliquot of 2 ml was collected and replaced by an equal report, a cholesterol level of 30 % mol/mol in Span: cho-
volume of the fresh release medium at the same tempera- lesterol niosomes of 7:3 was sufficient to stabilize niosomal
ture. The samples were analyzed for TM spectrophoto- bilayer membranes and imparting a residual gel/liquid
metrically at 295 nm. The kinetics of drug release was transition enthalpy a property known as thermo-respon-
studied by fitting the release data through three common siveness (Abdelkader et al. 2010), whereas a cholesterol
mathematical models (zero-order, first-order and Higuchi concentration of 50 % mol/mol (1:1 niosomes) was capa-
diffusion models). ble of abolishing gel/liquid transition of the bilayer mem-
branes (Uchegbu and Florence 1995; Abdelkader et al.
Ocular bioavailability study 2010). It is worthy to mention that TM has no appreciable
effects on the gel/liquid transition temperature of niosomes,
Pigmented rabbits, weighing between 1.0 and 1.5 kg, were as TM is a water soluble drug and preferentially encapsu-
employed in the experiments. The rabbits were fed bal- lated in the aqueous compartments of niosomes without
anced diet and maintained on 12 h/12 h light/dark cycle in affecting the gel/liquid transition of niosomes.
an air-conditioned room, at 28 C before the experiment.
The experimental procedures conformed to the ethical Entrapment efficiency (EE %) of TM
guidelines of Minia University Animal Ethics Committee
(Minia, Egypt). Effect of surfactant type
The rabbits were divided into five groups, 5 each, and
the basal IOP was measured for both right and left eyes of The EE % values for the prepared niosomes ranged from
all rabbits to give a basal IOP value at time zero using a 30 % 1.5 to 65 % 3 (Table 2). Span 40 and Span 60
pre-calibrated Schoetz tonometer (Riester, Germany). established the highest EE %, whereas the lowest EE %
An instilled dose of 2025 ll of TM solution or noisome values were recorded for Span 20. The main factors
dispersions, equivalent to TM concentration of 0.25 % w/v, affecting the entrapment of small water-soluble drug mol-
was instilled into the right eye surface of the rabbit, ecules in niosomes are the permeability of the bimolecular
whereas the same dose of the tested niosomes were membranes and the structure continuity of the hydrocarbon
instilled into the left eyes, so that the five groups are shown chain of the bilayer-forming surfactant (Grant et al. 2001).
in Table 1. Span 40 and Span 60 have long saturated acyl chains
Pharmacokinetic parameters such as Tmax and % [palmityl (C-16) and stearyl (C-18) chains respectively]
maximum reduction of IOP (IOPmax) were determined for which are in a gel state, compared with a liquid surfactant
the tested niosomes and the control TM solution. of short acyl chains [lauryl (C12)] at ambient conditions
Tmax is the time to maximum reduction in IOP (IOPmax). (Grant et al. 2001). Such features render Span 40- and Span
60-based niosomes less likely to leak the encapsulated
% maximum reduction of IOP %IOPmax
water-soluble drug molecules (TM). In contrast, Span
IOP0  IOPmax
20-based niosomes are fluid at room temperature and, even
IOP0
in the presence of high levels of cholesterol, are disorga-
where IOP0 is the intra-ocular pressure at 0 time, IOPmax is nized due to the trans-gauche conformations of their acyl
the intra-ocular pressure at Tmax. chains (Israelachvili et al. 1980). The EE % values for the

123
H. Abdelkader et al.

prepared niosomes can be summarized as follows: Span drug payload, compared with submicro-sized niosomes
60 [ Span 40 [ Span 20. These results support the (Uchegbu and Vyas 1998; Abdelkader and Alany 2012).
hypothesis that the higher the transition temperature of the
surfactant, the higher the EE % recorded for niosomes Effect of surfactant type
(Yoshioka and Florence 1994; Abdelkader et al. 2010).
The diameter values of Span 60-based niosomes were
significantly smaller than both Span 40-based and Span
Effect of cholesterol concentrations
20-based ones. Lipid aggregation and self-assembly in an
aqueous medium is mainly controlled by two opposing
Table 2 shows the effects of different cholesterol levels
electrostatic forces. These are hydrophobic attraction for-
used on EE %. Irrespective of surfactant type, the higher
ces between the hydrocarbon tail chains and electrostatic
the concentration of cholesterol incorporated into nio-
repulsion of the polar head groups (Tanford 1973). The
somes, the lower the EE % attained. This effect was
surface free energy increases with any decrease in hydro-
significantly recorded with Span 20-based niosmes
phobicity (Yoshioka and Florence 1994). Hence, surfac-
(shortest hydrophobic chain length); EE % for Span 20
tants of higher hydrophilicity (higher HLB) should yield
niosomes of cholesterol levels 30 and 50 % mol/mol was
larger vesicles.
45 and 30 % w/w respectively. Increases in cholesterol
concentration could increase the rigidity of the bilayer
Effect of cholesterol levels
membranes, by virtue of cholesterols rigid structure and
characteristic inverted cone shape (Israelachvili et al.
The increases in the mean diameter values were insignifi-
1980). This results in disruption of the vesicles bilayer
cant (P [ 0.05), when cholesterol level ranged from 30 %
structure which compromises their ability to entrap TM
(7:3) to 50 % (1:1) mol/mol. Similar results were reported
(Manosroi et al. 2003). This could lead to a decrease in
elsewhere (Essa 2010). Cholesterol is a rigid molecule with
the total number of bilayer vesicles available for encap-
an inverted cone shape. It can be intercalated between the
sulating the drug. Consequently, there is a decrease in the
fluid hydrocarbon chains of the amphiphile when hydrated
ability of the niosomes to entrap such water-soluble drug
at a temperature above the gel/liquid transition tempera-
molecules.
ture. It can also produce larger vesicles (Uchegbu and
Florence 1995; Essa 2010). Cholesterol is a membrane
Niosomal sizes stabilizer and can be accommodated and work coopera-
tively with the surfactant bilayer membranes.
Table 2 shows the mean volume diameter of the prepared
niosomes composed of various surfactants and contained Transmission electron microscopy (TEM)
two cholesterol levels (30 and 50 % mol/mol). The mean
diameter values of the prepared niosomes ranged from The microstructures of the prepared niosomes were studied
5.0 0.01 to 9.67 0.01 lm. The size measurements using TEM (Fig. 1). The micrographs captured revealed
were repeatable and reproducible from batch to batch spherical-shaped niosomes of smooth surfaces. However,
(acceptable deviations from the average values). These the diameters were not correlated with those obtained from
sizes are well accepted for ocular administration onto laser diffraction measurements.
surface of the eye, niosomal sizes of [10 lm were rec- The steps involved in the sample preparation (e.g. dur-
ommended for ocular delivery especially for water soluble ing replica formation on the copper grid and staining) for
drugs for better precorneal residence time and significant TEM are likely to promote the formation of smaller sizes

Table 2 Physical properties estimated for the prepared niosomes


Surfactant:cholesterol Ratio (mol/mol) Cholesterol level (%mol/mol) EE % w/w Volume diameter (lm) T3h (% w/w)

Control (PBS) 100 1.3


Span 20:cholesterol 7:3 30 45 2.3 5.0 0.01 68 2.0
1:1 50 30 1.5 11 .2 61 4.0
Span 40:cholesterol 7:3 30 58 2.2 4.9 0.03 50 1.7
1:1 50 52 2.0 8.0 0.01 36 4.7
Span 60:cholesterol 7:3 30 65 3.0 4.8 0.02 51 4.0
1:1 50 56 1.4 9.67 0.01 36 6.0

123
Effects of surfactant type and cholesterol level on niosomes

and artifacts during sample preparation and replica creation undergoing trans-gauch conformation all the time, whereas
(Egelhaaf et al. 2003; Perrie et al. 2007). Span 40 and Span 60 are gel and their hydrocarbon chains
are under restricted rotation. Accordingly, the encapsulated
In vitro release studies small drug molecules are easily partitioning out from fluid
bilayer membranes than gelled ones.
The in vitro release characteristics of TM from the pre- Cholesterol can be considered as mortar for niosomal
pared niosomes were studied on in-house designed diffu- bilayer membranes. Increasing the concentration of cho-
sion cells as mentioned in the method section. An aqueous lesterol is always associated with decreasing the perme-
solution of PBS was used as a release medium, stirred and ability of the encapsulated water soluble drug molecules
equilibrated at 35 C 0.5. The tested niosomes in the and release rate.
donor compartment were separated, by a cellulose mem- The TM release data obtained from the prepared nio-
brane, from the receptor compartment that was filled with somes were fitted into three mathematical models (zero-
20 ml of the release medium. order, first-order and Higuchi diffusion models) in order to
Figure 2 shows various release profiles of TM from the elucidate the best fitting model which can predict the
aqueous TM solution and the prepared niosomes. Per- mechanism of TM release. Table 3 summarizes the corre-
centage TM released at 3 h was denoted asT3h; the esti- lation coefficients (r) and release rate constants (k) for the
mated T3h for TM solution was 100 %, whereas that for all prepared niosomes. With the exception of Span 60-based
other niosomal formulations was significantly lower niosomes, it was obvious that the highest r values were
(P \ 0.05). This could be ascribed to encapsulating the recorded for the Higuchis model indicating that the best
water soluble molecules within the aqueous compartments fitting model was Higuchis and the general release
of niosomes. Accordingly, more time was required for TM mechanism for TM from the prepared niosomes was the
molecules to partition out of the bilayer membranes. These diffusion throughout the bilayer membranes of the
results indicated that niosomes can successfully control uncharged conventional niosomes. On the other hand, the
TM release. drug release from Span 60-based niosomes was dependent
With regard to effect of surfactant type, the values of on time rather than square root of time. These results are in
T3h calculated for Span 20-, Span 40- and Span 60-based a good agreement with those reported elsewhere (Guinedi
niosomes (7:3) were 68, 50 and 51 % respectively. These et al. 2005; Bayindir and Yuksel 2010). Further, the release
results could be attributed to the gel/liquid transition tem- rate constants calculated for the TM solution and the tested
perature (Tm) of niosomes forming surfactant. Span 20 niosomes were in a good agreement with the above men-
molecules are always fluid and their hydrocarbon chains tioned in vitro release parameter (T3h) suggesting that

Fig. 1 Micrographs of niosomes composed of Span 60: cholesterol 1:1 molar ratio

123
H. Abdelkader et al.

Fig. 2 In vitro release profiles


of timolol maleate (TM) from
TM solution and different
niosomal formulations

Table 3 Release kinetics parameters calculated for TM solution and niosomes


Formulation Zero order First order Higuchi
-1 -1
r K (%.h ) r K (h ) r K (%.h-0.5)

Control 0.975 36.40 0.915 1.40 0.990 7.64


Span 20:cholesterol 7:3 0.940 19.15 0.927 0.57 0.992 5.65
Span 20:cholesterol 1:1 0.942 17.22 0.974 0.37 0.990 5.07
Span 40:cholesterol 7:3 0.968 15.10 0.978 0.27 0.983 4.42
Span 40:cholesterol 1:1 0.954 10.58 0.984 0.15 0.988 3.11
Span 60:cholesterol 7:3 0.957 13.94 0.998 0.22 0.988 4.10
Span 60:cholesterol 1:1 0.964 10.26 0.993 0.14 0.987 3.00
r correlation coefficient, k release rate constant
Underlined data indicate the best fitting release mechanisms

niosomes successfully controlled the TM release and this Effect of surfactant type
effect is significantly dependent on the type of the surfac-
tant used and the cholesterol level in the bilayer Figure 4 displays the effect of surfactant type on the IOP
membranes. lowering profiles for the prepared niosomes. The values of
IOPmax were in the following order Span 40 [ Span
Ocular bioavailability study 20 [ Span 60. This behavior is not correlated with the
in vitro release profiles (Span 60 [ Span 40 [ Span 20).
Figure 3 shows the IOP-time curves for TM solution and Further, it could not be explained due to noisome size
niosomes composed of Span 20, Span 40 and Span 60. differences, as there were no significant differences in the
The time to trough IOP is called Tmax which is the time mean diameters for Span 20 7:3, Span 40 7:3 and Span 60
for maximum reduction of IOP (IOPmax). Tmax for TM 7:3 (Table 2). Other plausible explanation should be sought
solution and niosomes were 2 and 3 h respectively. In such as gel/liquid transition and the drug cargo capacity of
general, niosomes exhibited significantly (P \ 0.05) more niosomes. Therefore, the superior efficacy of Span
sustained ([7 h) reduction of the IOP compared with that 40-based niosomes could be accordingly explained. On one
for TM solution (less than 5 h). Also, the extent of IOP hand, the transition temperature (Tm) of Span 40 (45 C) is
reduction (as seen from the area under the curve, AUC) is less than that for Span 60 (55 C), this could impart rela-
significantly greater than that for TM solution. This is tively more flexibility of the hydrophobic bilayer mem-
evident from maximum percentage reduction of IOP branes, compared with that for Span 60-based niosomes.
(IOPmax) (Fig. 3). On the other hand, Span 40-based niosomes have

123
Effects of surfactant type and cholesterol level on niosomes

Fig. 3 Effect of surfactant type


on the ocular bioavailability
(IOP-time AUC) of Span-based
niosomes

Fig. 4 Summary of the effects


timolol maleate (TM) niosomes
of different surfactant types and
cholesterol concentrations on
the maximum percentage
reduction of IOP (IOPmax).
*P \ 0.05 indicates significant
differences. **P [ 0.05
indicates no significant
differences

Fig. 5 Representative graphs


for the effect of cholesterol
levels on ocular bioavailability
(IOP-time AUC) form Span
40-based niosomes

significantly greater capacity of the drug cargo than Span compromise between sufficient thermo-responsiveness and
20-based niosomes. Therefore, the highest IOPmax for Span capacity to load an efficient drug cargo, compared with
40-based niosomes could be attributed to having a good Span 20- and Span 60-based niosomes.

123
H. Abdelkader et al.

Effect of cholesterol levels Abdelkader H, Alani AW, Alany RG (2013) Recent advances in non-
ionic surfactant vesicles (niosomes): self-assembly, fabrication,
characterization, drug delivery applications and limitations.
Figure 5 shows representative curves of the effect of cho- Drug Deliv 21:87
lesterol concentrations on the IOP lowering activity for Aggarwal D, Garg A, Kaur IP (2004) Development of a topical
niosomes composed of Span 40: cholesterol at 7:3 and niosomal preparation of acetazolamide: preparation and evalu-
1:1 mol/mol. The AUC and IOPmax for Span 40 7: 3 nio- ation. J Pharm Pharmacol 56:15091517
Azmin MN, Florence AT, Handjani-vila RM, Stuart JFB, Vanlerr-
somes were significantly higher than those for Span 40 1:1. berghe G, Whittaker JS (1985) The effect of non-ionic surfactant
Cholesterol at a molar ratio of 30 % mol/mol imparts a vesicle (niosome) entrapment on the absorption and distribution
residual gel liquid transition which is likely to allow nio- of methotrexate in mice. J Pharm Pharmacol 37:237242
somes to transit from gel to fluid at ocular surface tem- Bayindir ZS, Yuksel N (2010) Characterization of niosomes prepared
with various nonionic surfactants for paclitaxel oral delivery.
perature (a property know as thermo-responsiveness), J Pharm Sci 99:20492060
whereas cholesterol concentration of 50 % mol/mol is Egelhaaf RM, Epand RF, Maekawa S (2003) The arrangement of
reported to completely abolish gel/liquid transition of cholesterol in membranes and binding of NAP-22. Chem Phys
niosomes. These finding support the hypothesis that a Lipids 122:3339
Essa AE (2010) Effect of formulation and processing variables on the
residual gel/liquid transition is favorably required for better particle size of sorbitan monopalmitate niosomes. Asian J Pharm
ocular drug uptake. 4:227233
Grant GJ, Barenholz Y, Piskoun B, Bansinath M, Turndorf H, Bolotin
EM (2001) DRV liposomal bupivacaine: preparation, character-
Conclusion ization, and in vivo evaluation in mice. Pharm Res 18:336343
Guinedi AS, Mortada ND, Mansour S, Hathout RM (2005) Prepa-
ration and evaluation of reverse-phase evaporation and multila-
Span-based niosomes (Span 20, Span 40 and Span 60) having mellar niosomes as ophthalmic carriers of acetazolamide. Int J
different gel/liquid transition temperatures of \10, 45 and Pharm 306:7182
55 C respectively, and two levels (30 and 50 % mol/mol) of Israelachvili JN, Marcelja S, Horn RG (1980) Physical principles of
cholesterol were prepared, characterized and in vivo evalu- membrane organization. Quart Rev Biophys 13:121200
Kassem MA, Abdel Rahman AA, Ghorab MM, Ahmed MB, Khalil
ated. It was found that both gel/liquid transition temperatures RM (2007) Nanosuspension as an ophthalmic delivery system
of the surfactants and the drug-cargo properties (EE %) of for certain glucocorticoid drugs. Int J Pharm 340:126133
niosomes contribute significantly to the ocular bioavailability. Kaur IP, Garg A, Aggarwal D (2004) Vesicular systems in ocular
This study can be considered a platform to study the influ- drug delivery: an overview. Int J Pharm 269:114
Kaur IP, Rana C, Singh H (2008) Development of effective ocular
ences of other parameters such size, the polar head group of preparations of antifungal agents. J Ocul Pharmacol Ther 24:
the surfactants and membranes additives on the ocular 481493
bioavailability. Kaur IP, Aggarwal D, Singh H (2010) Improved ocular absorption
kinetics of timolol maleate loaded into a bioadhesive niosomal
Acknowledgments This article dose not contain any studies with delivery system. Graefes Arch Clin Exp Ophthalmol 248:
human and animal subjects performed by any of the authors. All 14671472
authors (H. Abdelkkader, U. Farghaly, H. Moharram) declare that Liu T, Guo R (2007) Investigation of PEG 6000/Tween 80/Span
they have no conflict of interest. The authors alone are responsible for 80/H2O niosome microstructure. Colloid Polym Sci 285:
the content and writing of this article. 711713
Manosroi A, Wongtrakul P, Manosroi J, Sakai H, Sugawara F, Yuasa
M, Abe M (2003) Characterization of vesicles prepared with
various non-ionic surfactants mixed with cholesterol. Colloids
References Surf B Biointerfaces 30:129138
Nasr M, Mansour S, Mortada ND, Elshamy AA (2008) Vesicular
(1995). GraphPad InStat version 3.00, GraphPad Software, Inc., San aceclofenac systems: a comparative study between liposomes
Diego, www.graphpad.com and niosomes. J Microencapsul 25:499512
Abdelbary G, El-gendy N (2008) Niosome-encapsulated gentamicin for Perrie Y, Mohammed AUR, Vangala A, McNeil S (2007) Environ-
ophthalmic controlled delivery. AAPS PharmSciTech 9:740747 mental scanning electron microscopy offers real-time morpho-
Abdelkader H, Alany R (2012) Controlled and continuous release logical analysis of liposomes and niosomes. J Liposome Res
ocular drug delivery systems: pros and cons. Curr Drug Deliv 17:2737
9:110 Sahin NO (2006) Niosomes as nanocarrier systems. In: Mozafari MR
Abdelkader H, Ismail S, Kamal A, Alany R (2010) Preparation of (ed) Nanomaterials and nanosystems for biomedical applica-
niosomes as an ocular delivery system for naltrexone hydro- tions. Springer, New York, pp 6781
chloride: physicochemical characterization. Pharmazie 65: Schenker H, Maloney S, Liss C, Gormley G, Hartenbaum D (1999)
811817 Patient preference, efficacy, and compliance with timolol
Abdelkader H, Ismail S, Kamal A, Wu Z, Al-Kassas R, Alany R Maleate ophthalmic gel-forming solution versus timolol maleate
(2012) Conjunctival and corneal tolerability assessment of ocular ophthalmic solution in patients with ocular hypertension or
naltrexone niosomes and their ingredients on the hens egg open-angle glaucoma. Clin Ther 21:138147
chorioallantoic membrane and excised bovine cornea models. Int Sweetman S (2009) Matindale: the complete drug reference. Phar-
J Pharm 432:110 maceutical Press, London

123
Effects of surfactant type and cholesterol level on niosomes

Tanford C (1973) The Hydrophobic effect: formation of micelles and Yoshioka TBS, Florence AT, (1994) Preparation and properties of
biological membranes. Wiley, New York vesicles (niosomes) of sorbitan monoesters (Span 20, Span 40,
Uchegbu IF, Florence AT (1995) Non-ionic surfactant vesicles Span 60 and Span 80) and a sorbitsan triester (Span 85). Int J
(niosomes): physical and pharmaceutical chemistry. Adv Colloid Pharm 105:16
Interface Sci 58:155
Uchegbu IF, Vyas SP (1998) Non-ionic surfactant based vesicles
(niosomes) in drug delivery. Int J Pharm 172:3370

123

You might also like