Free Essay

Niosomal

In:

Submitted By Deepakhooda
Words 7354
Pages 30
FORMULATION AND EVALUATION OF TOPICAL GEL CONTAINING AZITHROMYCIN AND PREDNISOLONE VESICLES FOR TREATING PSORIASIS
Sonia Tomar, Tinku Singhal,

ABSTRACT
Psoriasis is a chronic, autoimmune systemic inflammatory disease, associated with metabolic syndrome, cerebrovascular disease, diabetes and many other diseases. There is various type of psoriasis but most common type of psoriasis is caused by Psoriasis vulgaris. It is characterized by rigid of skin due to increase in the level of cholesterol and fall in the level of ceramide. Apart from that it is associated with an immune system of the body means movement of immune cells from dermis to the epidermis, where they stimulate skin cells (keratinocytes) to proliferate. Various type of drug delivery system are used for the treatment of psoriasis including topical, oral or systemic but gels prepration of azithromycin and prednisolone are more effective in reduction of purities, scaling and hyperkeratosis of psoriasis plaque.
Niosomal/Vesicular gel, has been explored extensively for topical application to enhance skin penetration as well as skin retention. Prednisolone and azithroycin together provide effective results in the treatment of psoriasis. Due to high entrapment efficiency and stability, gel prepration (Azithromycin & Prednisolone) reduce the scaly patches and suppression of humoral immunity.
Keywords: Niosome, Immunity, Topical, Psoriasis, Gel, Azithromycin, Prednisolone.

INTRODUCTION
Psoriasis is recognized as a complex, chronic skin condition that can have a significant impact on patient’s physical and mental health. It occurs when the immune system sends out faulty signals that speed up the growth cycle of skin. It is characterized by rigid of skin due to increase in the level of cholesterol and fall in the level of ceramide (Surver C et al 2002). The main causes of the disease is not understand till now date but researchers correlate this disease with an immune system of the body means movement of immune cells from dermis to the epidermis, where they stimulate skin cells (keratinocytes) to proliferate (Franlk et al; 2009) and trigger the release of cytokinin (tumour necrosis factor- alpha TNF-α) which causes inflammation and the rapid production of skin cells (Asadullah et al; 1999). For the correction of psoriasis, a number of non drugs or salt are used like lithium, β-blockers and antimalarial (cholroquinine) drugs (James et al; 2005). Various Drug delivery systems are used for the treatment of psoriasis such as oral (Willms et al; 2010), systemic (Bremmer et al; 2010) & topically includes creams (Mesa et al; 2010), ointment (Gold et al; 2009), shampoo (Kircik et al;2010) & gel (Iraji et al; 2010). On the basis of effectivity in reduction of purities, scaling and hyperkeratosis of psoriasis plaques we select gels. Niosomes are composed of bilayer non-ionic surface active agents (Patel et al; 2007) consist of cholesterol and non ionic surfactant (Arora et al; 2007). They can be prepared by various types of methods like Ether injection method, Hand shaking method (thin film hydration techniques), Sonication, Microfluidization, Multiple membarane extrusion method ,Reverse phase evaporation technique (REV),
Trans membrane PH gradient (inside acidic ) drug uptake process, The bubble method, Formation of niosomes from proniosomes
Gel containing prednisolone & azithromycin are used for the treatment of psoriasis. As azithromycin as an antibiotic act against bacterial action which is one of the cause for the occurance for psoriasis & is also having keratolytic action to remove the scales of diseased skin. Prednisolone inhibit leukocyte infiltration at the site of inflammation, interfere in the function of mediators of inflammatory response, leads to suppression of humoral immune responses, and reduction in edema or scar tissue. The anti-inflammatory actions of corticosteroids are thought to involve phospholipase A2 inhibitory proteins, lipocortins, which control the biosynthesis of potent mediators of inflammation such as prostaglandins and leukotrienes. Niosomal gel provide the rapid penetration of drug through skin, restrict the action to target cell & act as a promising vehicle for the drug delivery by improving therapeutic performance of drug molecule, provide protection to drug from biological environment. Gel contain niosomes of prednisolone & azithromycin as sustained release of drugs are required. As half- life of azithromycin is too high of 68 hours to give a sustain release so there is no need to incorporate it in niosomes, hence directly incorporated in gel & prednisolone with short half life of 1 hour need to incorporate in noisome.

MATERIALS AND METHOD
Material
Chemical: Chloroform, Diethyl ether, Glycerin, Hydrochloric acid, Pottasium di hydrogen phosphate, sodium hydroxide, potassium chloride were purchased from Loba Chemic Pvt. Ltd., Central Drug House Pvt. Ltd.,
Surfactants: Cholesterol, Span 60, Span 40, Triton X 100 were purchased from S.D Fine Chem Ltd
Niosomes were prepared using ether injection method (Karki et al; 2008) by slowly introducing a solution of cholesterol and surfactant dissolved in diethyl ether into a warm phosphate buffer maintained at 600C. The surfactant mixture in ether was injected through guage needle 14 into an aqueous solution of drug. Vaporization of ether leads to formation of vesicles. Formulae for various niosomal formulations on the basis of varying concentration of surfactant and cholesterol are shown in table1.
Table1. Different formulation of noisome with varying concentration of surfactant Formulation | Surfactant (Span 60mg) | Cholesterol(mg) | Drug (mg) | Formulation | Surfactant (Span 60mg) | Cholesterol | Drug (mg) | F1 | 10 | 5 | 25 | F6 | 17 | 15 | 25 | F2 | 12 | 7 | 25 | F7 | 20 | 17 | 25 | F3 | 12 | 12 | 25 | F8 | 20 | 20 | 25 | F4 | 15 | 12 | 25 | F9 | 25 | 25 | 25 | F5 | 15 | 15 | 25 | 10 | 25 | 25 | 25 |

EVALUATION OF NIOSOMES
Microscopy
Optical microscopy :The microscopy of prepared niosomes was done by optical microscope. The niosomal suspension was placed over glass slide & observed for the formation of niosomes. The photomicrographs of the preparation were obtained with the help of photomicroscope (Kyowa Gatner 10390).
TEM (Transmission electron microscophy):Transmission electron microscopy was done to examine the size & morphology of prepared niosomes .A drop of dispersion was stratified into a carbon coated copper grid & left to adhere on the carbon substrate for about 1 minute. The dispersion in excess was removed by a piece of filter paper. The sample was air dried and observed under Hitachi transmission electron microscope at a voltage of 80 Kv (Muzzalupo et al; 2008).
Drug entrapment efficiency of niosomes
Entrapment efficiency of niosomes was determined by ultracentrifugation method. The dispersion was each ultracentrifuged at 12,000 rpm for 1 hr at 00 C. The supernatant was removed and formed niosomal pellet were resuspended in phosphate buffer solution pH (6.5) to ensure complete removal of all free Prednisolone. The supernatant (free Prednisolone) was collected and measured spectrometrically at λmax 245 using PBS as blank. The concentration of entrapped drug was determined spectrophotometrically after lysis of niosomal pellet with alcohol at 245 nm. Diethyl ethercontaining surfactant & cholesterol in the same ratio is used as blank in niosomal formulation. The entrapped concentration was expressed as % age entrapment efficiency which can be defined as the % age fraction of the total input drug encapsulated in the surfactant bilayer & aqueous compartment in the niosomes. It was calculated using following formulae % Entrapped efficiency = Entrapped Prednisolone ×100 Total drug (supernatant + sediment) In vitro release study The study was done by using membrane diffusion technique. In this 10 mg of niosomal formulation was placed in glass tube having diameter of 2.5 cm with an effective length of 8 cm that was previously covered with soaked osmosis cellulose membrane, which act as donar compartment. The glass tube was placed in a beaker containing 100 ml of phosphate buffer saline (pH 6.5). The receptor compartment containing suspension was just touching the surface of diffusion medium. The temperature of medium was maintained at 37±0.50 C and agitated at 100 rpm speed using magnetic stirrer. Aliquots of 5 ml of was withdrawn at different interval of time 0.5,1,2,3 up to 24 hours & same volume of medium was replaced with fresh medium. The collected samples were analyzed at 245 nm in double beam UV-Visible spectrometer using phosphate buffer (pH 6.5) as blank. Analysis of release mechanism
To ascertain the drug release mechanism, release rate of the niosomal formulations, data obtained from release studies of various niosomal formulation was fitted into various release models. The model selected were Zero order, First order, Higuchi, Korsemayer peppas model (Attia et al; 2007). The standard data for the interpretation of diffusional release mechanism is shown in table 2
Table2. Interpretation of diffusional release mechanism Release exponent | Drug transport mechanism | Rate as a function of time | n=0 | Fickian diffusion | t0.5 | 0.5>n>1 | Anomalous transport | tn-1 | n>1 | Case 2nd transport | Zero order releasetn-1 |

Zeta potential analysis
Zeta potential of niosomal preparation is related to the stability of niosomes. Zeta potential indicate the degree of repulsion between adjacent similarly charged particles for small molecules. High value of zeta potential confirms stability i.e the solution or dispersion will resist aggregation. Zeta potential for niosomal formulation was performed using zeta sizer. (Beckman coulter instrument) (Muuzzalupo et al.,2008).
Table3. Zeta potential value & corresponding stability Zeta- potential | Stability behavior | 0 to ± 5 | Rapid coagulation | ±10 to ± 30 | Incipient stability | ±30 to ± 40 | Moderate stability | ±40 to ±60 | Good stability | ±61 | Excellent stability |

Formulation of gel
Niosomal formulations exhibiting maximum in vitro release rate and high entrapment efficiency was selected for the formulation of gel containing different concentration of carbopol. AZI was added directly into gel base. Ingredients along with their quantities used in formulation of gel are shown in table 4

Table 4: Ingredients & quantities used in formulation of carbopol gel Ingredients | Quantity(F1) (50 gm) | Quantity(F2) (50 gm) | Quantity(F3) (50 gm) | Carbopol 934 | 500mg | 550mg | 450mg | Triethanolamine | 5 ml | 5ml | 5ml | Propylene glycol | 15ml | 15ml | 15ml | Glycerin | 0.845 ml | 0.845ml | 0.845ml | Prednisolone (niosomal pellet) | 25mg | 25mg | 25mg | Azithromycin | 100 mg | 100mg | 100mg | Methyl paraben | 25 mg | 25mg | 25mg | Propyl paraben | 250 mg | 250mg | 250 mg | Distilled water | Make up to 50 ml | Make up to 50 ml | Make up to 50 ml |

Parabens were dissolved in 40 ml of water with the aid of heat and allowed to cool. Carbopol 934 was added in small amount to the solution using high speed mixer until a smooth dispersion was obtained. To the dispersion glycerin, propylene glycol and known amount of drugs (PRE niosomal pellet, AZI) were added. Then neutralizing agent triethanolamine was added very slowly to avoid entrapped air. Finally, remaining water was added along with continous trituration to make a transparent gel.

EVALUATION OF GEL
Physiochemical evaluation of gel
Homogenity: All developed gel was tested for homogeneity by visual inspection after the gel has been set in the container. They were tested for their appearance and presence of aggregate.
Grittness: All the formulation was evaluated microscopically for the presence of particles if any.
Drug content studies
1.0 gm of each gel formulation were taken in 100 ml of volumetric flask containing 20 ml of phosphate buffer saline (pH 6.5) and stirred for 30 minute. The volume was made upto 100 ml & 1ml of the above solution was further diluted to 50 ml with phosphate buffer saline (pH 6.5). The resultant solution was filtered through membrane filter. The absorbance was recorded by using UV spectrophotometer at respective absorption maxima of AZI & PRE i.e 298 nm & 245 respectively. Drug content was determined from the calibration curve. pH determination
2.5 gm of gel was accurately weighed & dispersed in 25 ml of purified water. The pH of the dispersion was measured using pH meter.
Spreadibility
For the determination of spreadibility excess of sample was applied in between 2 glass slide & was compressed to uniform thickness by placing 1000 gm wt for 5 minute. Weight 50 gm was added to pan (Aejeaz et al; 2010). Time required toseparate the two slides i.e the time in which the upper glass slide move over the lower plate was taken as measure of spreadibility.
S= (m×l)/t
S= Spreadibility m = Weight tied to upper slide. l = Length moved on upper glass slide t = Time taken
Viscosity measurement
Viscosity of prepared formulation was determined using Brook field viscometer with spindle no 64 at 10 -50 rpm at temperature 37±0.50C (Martinez et al; 2007). Spindle was lowered perpendicularly into gel placed in a beaker taking care that the spindle does not touch the bottom of beaker . The spindle was rotated at different speed & reading were recorded after 30sec when the gel level stabilized. In vitro release permeation study of prepared gel
In vitro release studies was carried out using franz diffusion cell with receptor compartment volume of 25 ml & an effective area of 2.54 cm2. Receptor solution composed of distilled water.
Phosphate buffer saline (pH 6.5) was added to the cell & temperature was maintained at 37±0.50 C and solution was stirred continuously at 600 rpm(Bhatia et al; 2004). Wister rat skin was mounted on the receptor compartment with stratum corneum slide facing donar compartment. The donar compartment was filled with 200 mg of prepared gel. At appropriate time intervals of 0.5, 1, 2, 3 & upto 24 hrs,5 ml of sample were withdrawn & immediately replaced by equal amount of receptor solution. The sample were analysed by UV spectrophotometer at respective absorbance maxima of 298 nm & 245 nm.
Release kinetic studies of gel
In order to study the exact mechanism of drug release from the gels, drug release data was analyzed according to zero, first order, Higuchi square root and Korsemeyer- Peppas equations. Most appropriate model was chosen on the basis of goodness of fit test (Schmolka et l; 1972).
In vitro skin retenation study
The amount of drug retained in the skin was determined by using skin samples employed in permeation studies. After completion of permeation experiment, skin mounted on the diffusion cell was removed. The skin was cleaned with cotton dipped in saline solution and blotted with tissue paper to remove the adhering formuation. Then, the skin sample was homogenized with 20 ml of chlorofoam:methanol (2:1 v/v) for the extraction of rug (Bhatia et al; 2004). The solution thus obtained was filtered and absorbance was measured using UV spectrophotometer at respective absorption maxima of AZI (298nm) & PRE (245nm). Stability studies of gel
The prepared gel formulation were packed in air tight container at three different temperature conditions i.e refrigeration temperature (4-80C), room temperature (25±20C) & oven (45±20C). The sample were withdrawn at different time interval over a period of one month & evaluated for physical appearance & drug content. (Gupta et al;2007, Amriutiya et al;2009).

RESULTS:
Preparation and evaluation of niosome
Prednisolone loaded niosomes were prepared by ether injection method with varying concentration of surfactant & cholestrol. Niosome prepared were spherical in shape & found to be unilamillar (single layer vesicle) as observed under optical microscope (Fig 1)

Fig 1: Optical micrographs of niosomes
Histograms (Fig 2) were plotted between particle size distribution and no. of particles, representing the vesicle size of niosome in different formulation which were found to be in the range of 5-30µm, larger deviation in vesicle size can be explained on the basis of varying injection, speed of surfactant, cholestrol solution, height of injection due to aggregation of some vesicle & variation in temperature.( Ijeoma et al., 1998)

Fig 2: Histogram showing vesicles particle size range v/s number of particles.
Transmission electron microscopy of formulation F3 and F8 (fig 3) concluded that niosomes were discrete and spherical in shape (Mouzametal et al., 2011) with unilamillar structure

Fig 3: TEM of formulation F3 & F8
ENTRAPMENT EFFICIENCY
Entrapment efficiency of different formulation (F1-,F10) were found to be 32%, 35%, 38%, 47%, 51%, 58%, 62%, 66.5%, 64%, 63.26% respectively.
Table 5: Entrapment efficiency of formulations(F1-F10) Formulation | Surfactant quantity (mg) | Cholestrol (mg) | % Entrapment efficiency* | F1 | 10 | 5 | 32±1.324 | F2 | 12 | 7 | 35±0.439 | F3 | 12 | 12 | 38±0.180 | F4 | 15 | 12 | 47±0.275 | F5 | 17 | 15 | 51±0.362 | F6 | 20 | 15 | 58±0.586 | F7 | 20 | 17 | 62±0.975 | F8 | 34 | 20 | 66.5±0.134 | F9 | 50 | 25 | 64±0.694 | F10 | 75 | 25 | 63.26±0.965 |

Fig 4: Entrapment efficiency v/s surfactant
From data it was concluded from fig 4 that with increase in amount of cholesterol & surfactant the entrapment efficiency goes on increasing up to surfactant & cholesterol ratio of 2:1 (F8) and again decrease with increase (F9 & F10).(Rangasamy et al .,2008).Decrease in amount of surfactant may be due to high viscous systems produced due to high concentration of surfactant which affect niosome structure (Ijeoma et al., 1998).

In Vitro Release
Table 6: Cumulative %age release of formulation (F8-F10) Time (hr) | % Cumulative release of F8 | % Cumulative release of F9 | % Cumulative release of F10 | 0 | 0 | 0 | 0 | 0.5 | 10.082±0.043 | 6.145±0.491 | 4.69±0.956 | 1 | 14.79±0.064 | 7.97±0.532 | 10.42±0.487 | 2 | 25.28±0.654 | 10.141±0.876 | 16.9±0.657 | 4 | 39.62±0.865 | 14.97±0.563 | 22.3±0.693 | 6 | 45.05±0.876 | 17.94±0.435 | 28.05±0.547 | 8 | 51.07±0.564 | 22.2±0.638 | 31.56±0.386 | 10 | 53.69±0.543 | 24.6±0.345 | 33.13±0.548 | 12 | 59.28±0.432 | 24.8±0.456 | 40.189±0.657 | 14 | 64.84±0.432 | 26.27±0.567 | 46.73±0.874 | 16 | 73.35±0.431 | 20.98±0.465 | 59.9±0.765 | 18 | 70.76±765 | 20.75±0.765 | 67.63±0.564 | 20 | 67.95±0.576 | 19.98±0.654 | 61.58±0.362 | 22 | 66.96±0.674 | 19.87±0.764 | 59.95±0.673 | 24 | 66.28±0.896 | 18.64±0.325 | 57.85±0.964 |

Fig 5: % age cumulative release of formulation (F8-F10)

The result showed the consistent in vitro release in formulation F8 , then in formulation F9 and F10 due to presence of non-ionic surfactants in optimized concentration, which act as permeation enhancer (Arora et al., 2007) or due to small size of vesicles which enhances the effective surface area for penetration of drug via skin and due to greater concentration of prednisolone drug in formulation F8 vesicle in comparison to formulation F9 & F10 (Agarwal et al., 2001)
Analysis release mechanism
To ascertain drug release mechanism and release rate , in vitro of prepared formulation were fitted for various release models. The models selected were zero order, first order, Higuchi, Korsemeyer-Peppas model. All formulation are best fitted for Korsemeyer- Peppas equation. All formulation revealed non-fickian diffusion because slope of all formulations Korsemeyer- Peppas equation is near one.
Table 7: Data for analysis of release mechanism %CPR (F8) | Log CPR(F8) | %CPR (F9) | Log CPR(F9) | %CPR (F10) | Log CPR (F10 | T | √T | LogT | 10.082±0.043 | 1.003 | 6.145 ± 0.491 | 0.788 | 4.69±0.956 | 0.671 | 0.5 | 0.707 | -0.301 | 14.79 ± 0.064 | 1.169 | 7.97 ± 0.532 | 0.901 | 10.42±0.487 | 1.017 | 1 | 1 | 0 | 25.28 ± 0.654 | 1.402 | 10.141 ± 0.876 | 1.006 | 16.9±0.657 | 1.227 | 2 | 1.414 | 0.301 | 39.62 ± 0.865 | 1.597 | 14.97 ± 0.563 | 1.175 | 22.3±0.693 | 1.348 | 4 | 2.000 | 0.602 | 45.05 ± 0.876 | 1.653 | 17.94 ± 0.435 | 1.253 | 28.05±0.547 | 1.447 | 6 | 2.449 | 0.778 | 51.07 ± 0.564 | 1.708 | 22.2 ± 0.638 | 1.346 | 31.56±0.386 | 1.499 | 8 | 2.828 | 0.903 | 53.69 ± 0.543 | 1.729 | 24.6 ± 0.345 | 1.3909 | 33.13±0.548 | 1.552 | 10 | 3.162 | 1 | 59.28 ± 0.432 | 1.772 | 24.8 ± 0.456 | 1.394 | 40.189±0.657 | 1.604 | 12 | 3.464 | 1.079 | 64.84 ± 0.432 | 1.811 | 26.27 ± 0.567 | 1.419 | 46.73±0.874 | 1.669 | 14 | 3.741 | 1.146 | 73.35 ± 0.431 | 1.865 | 20.98 ±0.465 | 1.321 | 59.9±0.765 | 1.777 | 16 | 4 | 1.204 | 70.76 ± 0.765 | 1.849 | 20.75±0.765 | 1.317 | 67.63±0.564 | 1.83 | 18 | 4.242. | 1.255 | 67.95 ± 0.576 | 1.832 | 19.98±0.654 | 1.300 | 61.58±0.362 | 1.789 | 20 | 4.472 | 1.301 | 66.96 ± 0.674 | 1.825 | 19.87±0.764 | 1.298 | 59.95±0.673 | 1.777 | 22 | 4.690 | 1.342 | 66.28 ± 0.896 | 1.821 | 18.64±0.325 | 1.270 | 57.85±0.964 | 1.762 | 24 | 4.898 | 1.380 | Table 8: Kinetic release data of different model for formulations Model | Slope(F8) | R2 value (F8) | Slope (F9) | R2 value (F9) | Slope (F10) | R2 value (F10) | Zero order | 16.16 | 0.938 | 1.261 | -0.25 | 3.128 | 0.853 | First order | 0.634 | 0.463 | 0.027 | 0.385 | 0.049 | 0.637 | Higuchi | 19.76 | 0.923 | 5.531 | 0.577 | 12.86 | 0.937 | Korsemeyer Peppas | 0.503 | 0.964 | 0.369 | 0.812 | 39.06 | 0.847 |
From the kinetic release data (Table) it was concluded that formulation F8 follow korsemeyer peppas model & indicate anomalous diffusion mechanism diffusion coupled with erosion. Formulation F9 follow korsemeyer peppas model & indicate fickian diffusion mechanism. Formulation F9 follow korsemeyer peppas model & indicate fickian diffusion mechanism. . Formulation F10 follow Higuchi model & indicate diffusion controlled mechanism.
Zeta potential analysis
The zeta potential of niosomal suspension recorded by Beckman coulter delsa of formulation F8 was found to be 25.90mV. as shown in fig indicate thate suspension is stable, devoid of agglomeration and evenly distributed.

Fig 6: Zeta Potential of formulation F8

Formulation & evaluation of niosomal gel
Formulation F8 of niosome loaded with prednisolone having maximum in vitro release rate and highest entrapment efficiency was selected in combination with simple base azithromycin to formulate niosomal gel. The gel was formulated using carbopol and evaluated for following parameters
Physicochemical evaluation
The various physicochemical parameter (Table: 9) like grittiness, homogeneity was checked by observing formulation under microscope .
Table: 9 Physicochemical properties of formulation (F1-F3) Formulation | Homogeneity | Grittiness | %Drug Content (azithromycin) | %Drug Content (Prednisolone) | pH | Spreadibility(g.cm/s) | F1 | +++ | - | 87.5±0.54 | 90±0.67 | 6.7±0.65 | 15.85±1.68 | F2 | ++ | - | 92.4±0.65 | 90.5±0.98 | 7.1±0.51 | 14.65±2.42 | F3 | +++ | - | 89.7±0.43 | 88.9±0.46 | 7.3±0.32 | 11.25±1.56 |
Excellent +++, Good++, Satisfactory +, No grittiness-

Percent drug content in all three formulation was in range 87.5-92.5%, indicating homogeneity. pH of all formulations was found neutral (6.7-7.3). The spreadibility was found to be in the range of 11.25-15.61g cm/s. (Table:10 ) depicts the viscosity measurement at different angular velocity.
Table 10: Viscosity at different angular velocity Angular velocity (Spindle 60) | Viscosity (cps) | | F1 | F2 | F3 | 10 rpm | 7800±7.63 | 8200±17.55 | 8600±10.40 | 20 rpm | 6600±7.64 | 7700±20.20 | 7850±10.48 | 30 rpm | 6170±9.78 | 7480±11.54 | 7650±5.46 | 40 rpm | 6402±3.21 | 7210±7.61 | 7360±6.54 | 50 rpm | 6180±8.95 | 6900±9.57 | 7125±10.57 |

Fig: 7 Velocity at different angular velocity
Study depicts that formulation exhibited pseudoplastic rheology, as evidenced by shear thinning on increase in shear stress with increased angular velocity.The viscosities of formulations were in following orders F1<F2<F3. The viscosity increased with increasing concentration of sodium alginate and carbopol. F3 showed the maximum viscosity, this is due to change in polymeric concentration.

In vitro permeation study
Data on comparative in vitro permeation studies of prepared formulations at different time interval (Table:11) and amount of azithromycin & prednisolone were determined.
Table 11: Cumulative % release of different formulation (F1-F3) Time (hr) | %Cumulative drug release of F1 | %Cumulative drug release of F2 | %Cumulative drug release of F3 | | Azithromycin | Prednisolone | Azithromycin | Prednisolone | Azithromycin | Prednisolone | 0 | 0 | 0 | 0 | 0 | 0 | 0 | 0.5 | 2.16±0.098 | 8.09±0.031 | 1.81±0.054 | 5.32±0.078 | 0.96±0.006 | 3.36±0.321 | 1 | 5.68±0.876 | 12.07±0.098 | 3.46±0.081 | 7.76±0.465 | 2.04±0.076 | 7.24±0.543 | 2 | 9.98±0.765 | 21.03±0.081 | 7.48±0.765 | 9.03±0.654 | 4.18±0.065 | 12.96±0.643 | 4 | 12.08±0.534 | 33.98±0.079 | 9.84±0.654 | 11.79±0.764 | 6.82±0.041 | 19.30±0.865 | 6 | 16.84±0.532 | 41.07±0.312 | 13.52±0.043 | 15.49±0.864 | 7.96±0.076 | 23.28±0.986 | 8 | 19.43±0.876 | 47.09±0.217 | 17.39±0.321 | 19.97±0.564 | 11.01±0.065 | 28.56±0.061 | 10 | 23.31±0.007 | 50.03±0.538 | 19.28±0.876 | 23.74±0.623 | 15.84±0.067 | 30.38±0.650 | 12 | 28.08±0.786 | 54.82±0.243 | 21.04±0.985 | 25.72±0.543 | 18.09±0.085 | 37.89±0.976 | 14 | 33.07±0.795 | 61.29±0.421 | 24.08±0.865 | 22.89±0.326 | 22.04±0.086 | 43.63±0.065 | 16 | 37.04±0.841 | 67.53±0.058 | 29.48±0.041 | 21.93±0.541 | 25.98±0.016 | 40.34±0.341 | 18 | 48.09±0.684 | 65.32±0.216 | 32.19±0.086 | 21.07±0.0.654 | 28.84±0.032 | 62.76±0.768 | 20 | 54.91±0.086 | 63.95±0.076 | 48.28±0.076 | 19.98±0.754 | 31.42±0.065 | 60.85±0.658 | 22 | 61.42±0.976 | 62.46±0.036 | 53.04±0.041 | 20.36±0.548 | 34.67±0.075 | 58.67±0.885 | 24 | 64.06±0.865 | 61.96±0.069 | 58.98±0.065 | 20.28±0.659 | 38.48±0.043 | 55.58±0.662 |

Fig8: In vitro release of (a)prednisolone In vitro release of (b) azithromycin
Release Kinetic studies of niosomal gel
The release mechanism of prednisolone in niosomal gel was diffusion controlled and was influenced by polymer added. It was verified with different kinetic model (Table 12) All three formulation follows korsmeyer's plot which indicate that there is anomalous diffusion or diffusion coupled with erosion.
Table 12: Release kinetics of azithromycin from gel Model | Slope(F1) | R2 value (F1) | Slope (F2) | R2 value (F2) | Slope (F3) | R2 value (F4) | Zero order | 3.548 | 0.610 | 16.38 | 0.643 | 2.598 | 0.954 | First order | 0.03 | 0.670 | 0.039 | 0.605 | 0.052 | 0.693 | Higuchi | 14.96 | 0.949 | 6.69 | 0.931 | 13.77 | 0.956 | Korsemeyer Peppas | 0.548 | 0.964 | 0.483 | 0.962 | 0.717 | 0.984 |

Release in case of azithromycin all three formulation revealed non-fickian diffusion as the slope of all formulation for korsemeyer -peppas was near one and release mechanism was not influenced by variable added
Table 13: Release kinetics of Prednisolone from gel Model | Slope(F1) | R2 value (F1) | Slope (F2) | R2 value (F2) | Slope (F3) | R2 value (F4) | Zero order | 2.575 | 0.982 | 2.207 | 0.947 | 1.579 | 0.996 | First order | 0.057 | 0.792 | 0.058 | 0.827 | 0.056 | 0.844 | Higuchi | 13.04 | 0.899 | 8.568 | 0.791 | 6.261 | 0.865 | Korsemeyer Peppas | 0.795 | 0.972 | 0.827 | 0.973 | 0.926 | 0.992 | In vitro skin retention sudy
The percentage drug content (azithromycin & prednisolone) in skin from various formulatio ( Table 14) it was concluded that formulation FI was highly retained in skin as compared to other two solution
Table 14: %age of drug retained on skin S. No. | Formulation | % drug retained (Azithromycin) | % drug retained (prednisolone) | 1 | F1 | 23±0.76 | 59±0.12 | 2 | F2 | 11±0.57 | 26±0.76 | 3 | F3 | 19±0.42 | 39±0.63 |

Stability studies
The stability studies were conducted on formulation F1 at different temperature & humidity condition. refrigeration and at room temperature up to one month showed that formulation was stable (Table15)
Table 15: Stability study of drus at different temperature at different time interval Weeks | RefrigerationAzithromycin | RefrigerationPrednisolone | At 25o CAzithromycin | At 25o CPrednisolone | At 45o CAzithromycin | At 45o CPrednisolone | 1 | 96.65±1.54 | 92.83±0.54 | 93.45±0.75 | 91.87±1.63 | 87.96±0.98 | 87.20±0.98 | 2 | 86.74±1.23 | 90.43±0.36 | 88.54±0.86 | 86.82±1.76 | 85.45±0.56 | 83.43±0.76 | 3 | 85.64±0.96 | 85.64±1.14 | 85.73±0.97 | 84.63±0.56 | 83.42±0.45 | 78.42±0.79 | 4 | 83.31±0.87 | 82.43±0.94 | 82.03±1.32 | 82.25±0.79 | 81.56±0.23 | 76.44±1.42 | 5 | 82.34±1.81 | 80.65±1.56 | 81.76±0.54 | 79.08±0.42 | 78.95±1.18 | 75.08±1.56 | * Each value represnt the mean ±S.D. of 3 determination Fig 9: Histogram Showing % of Azithromycin and Prednisolone retained at various temperature

CONCLUSION

Niosomal/Vesicular gel, has been explored extensively for topical application to enhance skin penetration as well as skin retention. Prednisolone and azithroycin together provide effective results in the treatment of psoriasis. Prednisolone was incorporated in vesicle using span 60 (hydrophilic span form vesicle & entrap lipophilic drug) as surfactant, which give high entrapment efficiency and high stability to prepared vesicles. Prednisolone help in reduction of scar by suppression of humeral immune response. Azithromycin is used to remove scaly patches and to avoid the effects produced by prednisolone. Entrapment efficiency were checked by centrifugation method and was found maximum in formulation F8 having surfactant & cholesterol in ratio 2:1. Niosomal prednisolone formulation were evaluated for parmaters like microcopy, particle size, shape, drug content, in vitro release, zeta potential etc . All formulation size range of 50-100µm. Drug release mechanism were confirmed to be non-fickian diffusion by following kossermeyer peppas kinetic model. Azithromycin was incorporated into simple base gel along with dried niosomal pellets of prednisolone.All formulation tend tend to have confirmatory physicochhemical properties as pH tend to neutral, viscosity varies by adding amount of carbopol in different amount. Gel tend to be easily spreadable as its value come in range of 11.2-15.6g.cm/sec. F1 formulation provide highest in vitro release rate. Drug release mechanismfrom gel were confirmed to be non-fickian diffusion by following kossermeyer peppas kinetic model. Stability study showed no significant change when kept under different temperature & humidity conditions.

REFERENCE

Aejaza, A., Azmail, K., Sanaullah, S., Mohsin, A.A., 2010. Studies of Aceclofenac solid dispersion incorporated gels. Development, characterization and in vitro evaluation. Int. J.Pharm. Sci. 2, 111-115.
Ali, J., Akhtar, N., Sultana, Y., Baboota, S., Ahuja, A., 2008. Antipsoriatic microemulsion gel formulations for topical drug delivery of babchi oil (Psoralea corylifolia). Exp. Clin. Pharmacol. 30, 277-85.
Ali, MF., Salah, M., Rafea, M., Saleh, N., 2008. Liposomal methotrexate hydrogel for treatment of localized psoriasis: preparation, characterization and laser targeting. Med. Sci. Monit. 14, 66-74.
Amrutiya, N., Baja, A., Madan, M., 2009. Development of microsponges for topical drug delivery of Mupirocin. AAPS. Pharm . Sci. 10, 402-409.
Andersen, S.M., Rosada, C., Hansen, F., Laugesen, I.G., Darko, E., Dam, T.N., Stenderup, K., 2010. Topical application of valrubicin has a beneficial effect on developing skin tumors. Carcinogen. 31(8), 1483-90.
Angelique, N., Karen, T., Jeffery, S., Daniel, F., 2008. Psoriatic skin lesions induced by tumor necrosis factor antagonist therapy. 59(7), 996–1001 Arora, R., Jain, C.P., 2007. Advances in noisome as a drug carrier: a review. Asin J. Pharm. 1 29-39. Attia, A., Gizaway, S.A., Fouda, M.A., Donia, A.M. 2007. Influence of niosomal formulation o the oral bioavailabilityof acyclovir in rabbit. AAPS. Pharm. Sci. Tech. 8 (4), 106 Azmin, M.N., Florence, A.T., Handjani, R.M., Stuart, J.F.B., Vanlerberghe, G., Whittaker, J.S., 1985. The effect of non-ionic surfactant vesicles (niosomes) entrapment on the absorption and distribution of methotrexate in mice. J. Pharm. Pharmacol. 37, 237-242. Baillie, A.J., Florence, A.T., Hume, L.R, Rogers, A., Muirhead, G.T., 1986. The preparation and properties of niosomes non-ionic surfactant vesicle. J. Pharm. Pharmacol. 37, 863-868.
Banker, G.B.S., Rodes, C.T., 1979. Transdermal drug delivery. Mod. Pharm. 40, 263-273.
Behnam, S.M., Behnam, S.E., Koo, J.Y., 2005. Smoking and psoriasis. Skinmed. 4, 174–6.
Bhatia, N., 2004. Tamoxifen in topical liposomes. Development, characterization and in vitro evaluation. J. Pharm. Pharmaceut. Sci. 7, 252-259. Chandraprakash, K.S., Udupa, N., Umadevi, P., Pillai, G.K., 1992. Formulation and evaluation of Methotrexate niosomes. Ind. J. Pharm. Sci. 54, 197. Chauhan, S., Luorence, M.J., 1989. The preparation of polyoxyethylene containing non-ionic surfactants vesicles. J. Pharm. Pharmacol. 41, 6. Cummings, J., Staurt, J.F., Calman, K.C., 1984. Determination of adriamycin, adriamycinol and their 7- deoxyaglycones in human serum by high-performance liquid chromatography. J.Chrom. 311, 125-133. Digun, B., Cengiz, E., Gokhan, H., Fusun, B., Dilek, B., 1999. Coexistence of morphea and psoriasis responding to acitretin treatment. J. Eur. Acad. Dermatol. Venereol. 13, 113-117.
Faiyazuddin, M., Akhtar, N., Akhter, J., Suri, S., Shakeel, F., Shafiq, S., Mustafa, G., 2010.
Production, characterization, in vitro and ex vivo studies of babchi oil-encapsulated nanostructured solid lipid carriers produced by a hot aqueous titration method. Pharmazie. 65(5), 348-55.
Fang, Y.P., Huang, Y.B., Wu, P.C., Tsai, Y.H., 2009. Topical delivery of 5-aminolevulinic acid-encapsulated ethosomes in a hyperproliferative skin animal model using the CLSM technique to evaluate the penetration behavior. Eur. J. Pharm. Biopharm. 73(3), 391-8.
Frank, O., Nestle, D.H.K., Barker, J., 2009. Mechanisms of Disease. Psoriasis. N. Engl. J. Med. 361, 496-509.
Freedberg, M., Fitzpatrick, B., 2003. Fitzpatrick's dermatology in general medicine . Mc Graw- Hill. 6, 414.
Gayatri, S., Venkatesh, P., Udupa, N., 2000. Niosomal sumatriptan succinate for nasal administration. Int. J. Pharm. Sci. 62, 479-481.
Gelfand, J.M., 2005. Prevalence and Treatment of Psoriasis in the United Kingdom. Arch. Dermatol. 141 (12) 1537–1541.
Gold, L.F., 2009. Calcitriol ointment: optimizing psoriasis therapy. J Drugs Dermatol. 8, 23-Gregoriadis, G., 1981. Targetting of drugs: implications in medicines. Lancet. 2, 241-246.
Gudjonsson, J.E., 2002. HLA-Cw6-positive and HLA-Cw6-negative patients with psoriasis vulgaris have distinct clinical features. J. Inves. Dermatol. 118(2), 362-365.
Gupta, A., Parajapati, S.K., Balamurugan, M., Singh, M., Bhatia, D., 2007. Design and development of proniosomal trandermal drug delivery system for Captopril. Trop. J. Pharm. Res. 6, 687-693.
Hao, Y., Zhao, F., Li, N., Yang, Y., 2002. Studies on a high encapsulation of colchicines by noisome system. Int. J. Pharm. 244, 73-80.
Huczko, A., Lange, H. F., 1999. Experimental evidence for a null risk of skin irritation and allergy. Fullerene Sci. Technol. 7, 935-939. Hu, C., Rhodes, D.G., 1999. Proniosomes: a novel drug carrier preparation . Int . J. Pharm. 185, 23-35. Ijeoma , F., 1998. Nonionic surfactant based vesicles in drug delivery. Int. J. Pharm. 172, 33-70.
Iraji, F., Faghihi, G., Siadat, AH., Enshaieh, S., Shahmoradi, Z., Joia, A., Soleimani, F., 2010. Efficacy of 15% azelaic acid in psoriasis vulgaris: a randomized, controlled clinical J. Drug. Dermatol. 9, 964-8.
Jacobi, A., Braeutigam, M., Mahler, V., Schultz, E., Hertl, M., 2008. Pimecrolimus 1% cream in the treatment of facial psoriasis. Dermatol. 216(2), 133-6 .
Jain, N.K., 1997. Controlled and novel drug delivery. CBS Publishers and Distributors, Delhi, 32, 100-106. Jayaraman, C.S., Ramachandran, C., Weiner, N., 1996. Topical delivery of erythromycin from various formulations: an in vivo hairless mouse study. J. Pharm. Sci. 85, 1082-1084.
Joar, A., Jens, R., Bjorn, T., John, K., 1998. Clobetasol propionate followed by calcipotriol is superior to calcipotriol alone in topical treatment of psoriasis. Journal of the Eur. Acad. Dermatol.Venereol. 11, 19-24.
Jones, S.E., Boyle, J., Harman, R.R.M., 1987. Local PUVA treatment for nail psoriasis. Br. J. Dermatol. 9116, 280–281.
Jong, E.M.G.J., Seegers, B.A.M.P.A., Gulinck, M.K., 1996. Psoriasis of the nails associated with disability in a large number of patient. Dermatol. 193, 300-303.
John., 1998. Mometasone furoate 0.1% salicylic acid 5% ointment versus mometasone furoate 0.1% ointment in the treatment of moderate-to-severe psoriasis. Clinc. Therap. 20, 2, 283-291.
Karki, R., Mamatha, G.C., Subramanya, G., Udupa, N., 2008. Preparation, characterization and tissue disposition of niosomes containing isoniazid. Rasayan. J. Chem. 1, 224-227. Khandare, J.N., Madhav, G., Tamhankar B.M., 1994. Niosomes as novel drug delivery system. East. Pharm. 37, 61-64.
Louden, B.A., Pearce, D.J., Lang., W, Feldman., 2004. A Simplified Psoriasis Area Severity Index (SPASI) for rating psoriasis severity in clinic patients. J. Dermatol. 10, 2- 7.
Lemberger, A.P., 1973.Cutaneous and transdermal delivery. A hand book of non prescription drug, Am. Pharma. Assoc., 23, 161.
Leo., 1994. Topical formulation development of a novel thymidylate synthase inhibitor for the treatment of psoriasis. Int. J. Pharm.105,3, 227-233.
Lin, Y.K., Huang, Z.R., Zhuo, R.Z., Fang, J.Y., 2010. Combination of calcipotriol and methotrexate in nanostructured lipid carriers for topical delivery. Int. J. Nanomed. 5, 117-
128.
Lakshmi, P.K., Devi, G.S., Bhaskaran, S., 2007. Niosomal methotrexate gel in the treatment of localized psoriasis Phase I and phase II studies. Int. J. Dermatol. Venereo. 3, 157-161.
Malhotra, M., Jain, N.K., 1994. Niosomes as drug carriers. Indian Drugs, 31,81-86.
Martini, F., & Nath, J., 2009. Fundamentals of Anatomy & Physiology Pearson Education. J. Dermatol. 8, 158.
Mithal, B.M., Saha, R.N., 2003. A handbook of cosmetics, Vallabh Prakashan Delhi. 1, 214-215.
Montazeri, F., Kanitakis, J., Bazex, J., 1996. Psoriasis and HIV infection. Int. J. Dermatol. 35, 475-479.
Morganti, P., Ruocco, E., Wolf, R., Ruocco, V., 2001. Percutaneous absorption and delivery systems. Clin. Dermatol. 19,489.
Manconi., 2002. Niosomes as carriers for tretinoin I: Preparation and properties. Int. J. Pharm. 234, 237-248.
Manconi., 2003. Niosomes as carriers for tretinoin II: Influence of vesicular incorporation on tretinoin photostability. Int. J. Pharm. 260, 261-272.
Manconi., 2006. Niosomes as carriers for tretinoin III: A study into the in vitro cutaneous delivery of vesicle-incorporated tretinoin. Int. J. Pharm. 311, 11-19. Maver, L.D., Bally, M.B., Hope, M.J., Cullis, P.R., 1985. Biochem. Biophys. Acta. 816, 294- 302.
Mesa, R.A., 2010. Ruxolitinib, a selective JAK1 and JAK2 inhibitor for the treatment of myeloproliferative neoplasms and psoriasis. J. Invest. Drug. 13, 2040-3410. Moser, P., Marchand, M., Labrude, P., Handjani, R.M., Vignerson, C., 1989. Hemoglobin niosomes. I. Preparation, physiochemical properties and oxygen- carrying capacity, stability . Pharma. Acta. Helv .64, 192-202. Mouzam, M.D., Mouzam, I., Dehghan, M.H.G., Shaikh, M., 2011. Development and characterization of salmetrol xinafoate for nasal delivery. Ind. J. Pharm. Edu Res, 45, 121-127. Mulik, R., Mahadik, K., Paradkar, A., 2009. Development of curcuminoids loaded poly(butyl) cyanoacrylate nanoparticles. Physicochemical characterization and stability study . Eur. J. Pharm. Sci. 37, 395-404. Naresh, R.A., Chandrashekhar, G., Pillai, G.K., Udupa, N., 1994. Antiinflammatory activity of niosome encapsulated diclofenac sodium with tween -85 in arthritic rats. Ind. J. Pharmacol. 26, 46-48. Osborne, D.W., Henke, J.J., 1997. Skin penetration enhancers cited in the technical literature. Pharm. Tech. 21, 50-66. Ping, F., Yaw, H., Pao, Wu., Hung, T., 2009. Topical delivery of 5-aminolevulinic acid- encapsulated ethosomes in a hyperproliferative skin animal model using the CLSM technique to evaluate the penetration behavior. Eur. J. Pharm. Sci.73, 391-398. Posthumd, J.J., Vink, J., Lecessies J.A., 1998. Topical Tretenoin under occlusion on a typical navei. Pharmaceutical dosage form and drug delivery system. 7, 548. Prausnitz, M.R., Bose, V.G., 1995. Electroporation: In percutaneous penetration enhancers, CRC Press, Bocaraton, 393-405. Rakesh, P., 2007. Niosome: An Unique Drug Delivery System.Pharma.info.net.5,6. Rangaswamy, M., Ayyasamy, B., Raju, S.K., Gummadevelly, S., Shaik, S., 2008. Formulation and in vitro evaluation of noisome encapsulated acyclovir. J. Pharm. Res. Rudolf, A., 2000. Guttate psoriasis triggered by perianal streptococcal dermatitis in a four-year- old boy. J. Am. Acad. Dermatol. 42,5, 885-887. Schmolka, I.S., 1972. Formulation and evaluation of ophthalmic gel for sustained release of drug. J. Biomed.M. Res. 6,571-75. Shah, V.P., Williams, R.L., 1993. Skin penetration enhancement. Clin. Pharmacol. 27-35. Shapiro, J., Cohen, AD., David, M., Hodak, E., Chodik, G., Viner, A., Kremer, E., Heymann, A. 2007. The association between psoriasis, diabetes mellitus, and atherosclerosis in Israel: a case-control study. J. Am. Acad. Dermatol. 56(4), 629-34. Stine, M.A., Cecilia, R., Hansen, F., Laugesen, I.G., 2010, Topical application of valrubicin has a beneficial effect on developing skin tumors. J. Oxd. 31, 1483-1490. Surver, C., Davis, F.A., 2002. Bioaviability and bioequivalence, In Walter, K.A: Dermatological and transdermal formulation, Marcel Dekker, INC. New York, 119. 403. Tardi, C., Brandl, M., Schubert, R., 1998. Erosion and controlled release properties of semisolid vesicular dispersion. J. Control. Release., 55, 261-270. Terech, P., 1997. Low-molecular weight organogelators. Specialist surfactants. Glasgow: Blackie. Academic and Professional. 208–268. Treloar, V., 2010. Integrative dermatology for psoriasis: facts and controversies. Clinic. Dermatol. 28 ,1, 93–99. Van, CM., 2003. Psoriasis. Dermatol. 125–149. Van, J., Schoonbeek, F., Loos, M., Veen, E.M., Kellog, RM., Feringa, B.L., 1999. Low molecular weight gelators for organic solvents. Supramolecular science. Kluwer Academic Publishers. 233–259. Vyas, S., Khar, R.K., 2002. Controlled drug delivery – Concept and advances, Vallabh Prakashan. 418-422. Weissman, G., Bloomgarden, D., Kaplan, R., Cohen, C., Hoffstein, S., Collins, T., Gotlieb, A.,1975. A general method for the introduction of enzymes, by means of immunoglobulin- coated liposomes, into lysosomes of deficient cells. Proc. Natl. Acad. Sci. 72, 88-92. Wolters, M., 2005. Diet and psoriasis: experimental data and clinical evidence. Br. J Dermatol.153, 4,706-14. Yoshida, H., Lehr, C.M., Kok, W., Junginger, H.E., Verhoef, J.C., Bouwistra, J.A., 1992.Niosomes for oral delivery of peptide drugs. J. Control Rel. 21, 145-153. Yoshioka, T.Y., Sternberg, B., Florence, A.T., 1994. Preparation and properties of vesicles (niosomes) of sorbitan monoester (span 20, span 40, span 60 and span 80) and a sorbitan trimester (span85). Int. J. Pharm. 105, 1-6. Yilmaz, E., Borchert, H.H., 2006. Effect of lipid-containing, positively charged nanoemulsions on skin hydration, elasticity and erythema--an in vivo study. Int. J. Pharm. 307, 2, 232-8. Walsh, A.I., Rhodes, D.G., 2001. SEM imaging predicts quality of niosomes from maltodextrin based proniosomes. Pharm. Res. 18, 656-661.
William, J., Timothy, B., Dirk, E., 2005. Andrews Diseases of the Skin. Clin. Dermatol. 10, 191– 7.
Zenz. R, Eferl R, Kenner L., 2005. Psoriasis-like skin disease and arthritis caused by inducible epidermal deletion of Jun proteins. Nat. 437, 369–75. Zhou, N., Bai, Y.P., Man, X.H., Zhang, YB., Kong, Y.H., Chang M., 2009. Effect of new Pulian ointment in treating psoriasis of blood-heat syndrome. Chin. J. Integr. Med. 15, 6,409- 14.

Similar Documents