Enhanced Dissolution Rate of Atorvastatin Calcium using Solid Dispersion with PEG 6000 by Dropping Method

Similar documents
Enhancement of dissolution rate of atorvastatin calcium using solid dispersions by dropping method

COMPARATIVE EVALUATION OF SOLUBILITY AND DISSOLUTION ENHANCEMENT OF BICALUTAMIDE SOLID BY DISPERSION TECHNIQUE

DISSOLUTION PROFILLING OF NIMESULIDE SOLID DISPERSIONS WITH POLYETHYLENE GLYCOL, TALC AND THEIR COMBINATIONS AS DISPERSION CARRIERS

FORMULATION AND EVALUATION OF REPAGLINIDE FAST DISSOLVING TABLETS

IMPROVEMENT OF DISSOLUTION PROFILE OF LORNOXICAM BY SOLID DISPERSION USING SPRAY DRYING TECHNIQUE

Solubility and Dissolution Rate Determination of Different Antiretroviral Drugs in Different ph Media Using UV Visible Spectrophotometer

Enhancement of Solubility and Dissolution of Celecoxib by Solid Dispersion Technique

Available online through ISSN

Ondansetron hydrochloride (OSH) is an effective

8. FORMULATION OF LANSOPRAZOLE NANOPARTICLES

INTERNATIONAL JOURNAL OF PHARMACEUTICAL AND CHEMICAL SCIENCES

Received 25 March, 2010; received in revised form 03 June, 2010; accepted 15 June, 2010

Preparation And Characterization Of Simvastatin Nanosuspension By Homogenization Method

Md.Khairul Alam et al / Journal of Pharmaceutical Science and Technology Vol. 3 (6), 2011,

FORMULATION AND EVALUATION OF FAST DISSOLVING CHLORTHALIDONE TABLETS

FORMULATION AND EVALUATION OF SOLID DISPERSIONS OF CARVEDILOL, A POORLY WATER SOLUBLE DRUG BY USING DIFFERENT POLYMERS

In Vivo-In Vitro Evaluation of Solid Dispersion Containing Ibuprofen

Formulation and Evaluation of Immediate Release Tablets of Nevirapine Solid Dispersions

Development of Discriminating Method for Dissolution of Aceclofenac Marketed Formulations

Asian Journal of Biomedical and Pharmaceutical Sciences 1 (3) 2011, 13-19

FOR IMPROVED BIOAVAILABILITY

DISSOLUTION ENHANCEMENT OF ACECLOFENAC SOLID DISPERSION PREPARED WITH HYDROPHILIC CARRIERS BY SOLVENT EVAPORATION METHOD

UV Spectrophotometric Method Development and Validation of Ezetimibe and Simvastatin in Bulk and Pharmaceutical Dosage Form

FORMULATION AND EVALUATION OF FAST DISSOLVING TABLETS OF GLICLAZIDE

Enhancing the solubility of Candesartan cilexetil-inclusion Complexation using

International Journal of Pharmacy and Pharmaceutical Sciences Vol 2, Issue 1, 2010

King Saud University College of Pharmacy Department of Pharmaceutics. Biopharmaceutics PHT 414. Laboratory Assignments 2010 G 1431 H

EFFECT OF POLOXAMER 188 ON IN VITRO DISSOLUTION PROPERTIES OF ANTIPSYCHOTIC SOLID DISPERSIONS

CHAPTER V ANALYTICAL METHODS ESTIMATION OF DICLOFENAC. Diclofenac (gift sample from M/s Micro Labs Ltd., Pondicherry)

Dissolution Method Development and Validation of Paracetamol Aceclofenac Tablets

EFFECT OF PVP K30 ON COMPLEXATION AND DISSOLUTION RATE OF NEVIRAPINE β CYCLODEXTRIN COMPLEXES

PHARMA SCIENCE MONITOR FORMULATION AND EVALUATION OF SOLID DISPERSION OF OLANZEPINE

Validated RP-HPLC Method for Estimation of Cefprozil in Tablet Dosage Form

Enhancement of Solubility and Dissolution Rate of Poorly Soluble Antihypertensive Drug using Solid Dispersion Technique

Dicyclomine-loaded Eudragit -based Microsponge with Potential for Colonic Delivery: Preparation and Characterization

Research Article. Dissolution Study of Oxolamine Citrate by UV Spectrophotometric Method in Pharmaceutical Dosage Form

104 Full Text Available On Research Article!!! Pharmaceutical Sciences. Received: ; Accepted:

Journal home page:

RESEARCH ARTICAL PREPARATION AND CHARACTERIZATION OF SELF EMULSIFYING DRUG DELIVERY SYSTEM OF TELMISARTAN

Chapter 7 FORMULATION AND CHARACTERIZATION OF PULSINCAP

Preparation, Characterization, and Evaluation of Physical Mixtures of Ibuprofen for Solubility Enhancement

International Journal of Pharma and Bio Sciences

Estimation of Dapagliflozin from its Tablet Formulation by UV-Spectrophotometry

Estimation of Fenofibric Acid in Pharmaceutical Oral Solid Dosage Form by UV-Spectrophotometry

Research Article DESIGN AND EVALUATION OF SUSTAINED RELEASE FORMULATIONS OF THEOPHYLLINE USING NATURAL POLYMERS

Revision Bulletin 27 Jan Feb 2017 Non-Botanical Dietary Supplements Compliance

DETERMINATION OF DRUG RELEASE DURING DISSOLUTION OF NICORANDIL IN TABLET DOSAGE FORM BY USING REVERSE PHASE HIGH PERFORMANCE LIQUID CHROMATOGRAPHY

Spectroscopic Method For Estimation of Atorvastatin Calcium in Tablet Dosage Form

Improvement of Dissolution Properties of Furosemide by Complexation with p=c yclodextrin

CHAPTER 7 SUMMARY AND CONCLUSIONS. constitutes the objective of many a research project in the field of pharmaceutical

DEVELOPMENT AND VALIDATION OF RP-HPLC METHOD TO DETERMINE CINITAPRIDE HYDROGEN TARTARATE IN BULK AND PHARMACEUTICAL FORMULATION

Dissolution study and method validation of alprazolam by high performance liquid chromatography method in pharmaceutical dosage form

Formulation and Characterization of Asenapine Maleate Nanoparticles

Revision Bulletin 29 Dec Jan 2018 Non-Botanical Dietary Supplements Compliance

Comparison of US Pharmacopeia Simulated Intestinal Fluid TS (without pancreatin)

Development and validation of septrophotometricmethods for the estimation of rasagiline in tablet doage form

IN VITRO DISSOLUTION KINETIC STUDY OF THEOPHYLLINE FROM HYDROPHILIC AND HYDROPHOBIC MATRICES

Pelagia Research Library

Plop Plop, Fizz Fizz, Oh What A Relief It Is (Which Pain Reliever Works Fastest)

AREA UNDER CURVE AND SECOND ORDER DERIVATIVE SPECTROSCOPY OF METAXALONE IN BULK DRUG AND TABLET FORMULATION

International Journal of PharmTech Research CODEN (USA): IJPRIF, ISSN: , ISSN(Online): Vol.9, No.7, pp , 2016

Available Online through

Validated spectrophotometric determination of Fenofibrate in formulation

Development and validation of UV- spectrophotometric method for the estimation of dabigatran etexilate mesylate (dem)

INTERNATIONAL JOURNAL OF PHARMACEUTICAL RESEARCH AND BIO-SCIENCE

Scholars Research Library. Innovation on Development and Evaluation of Gastric Oral Floating Capsules Containing Captopril

Research Article Spectrophotometric Estimation of Didanosine in Bulk Drug and its Formulation

Dr Mingzhong Li School of Pharmacy, De Montfort University

Practical Pharmaceutical Technology I USP Dissolution Method for PARACETAMOL 500 mg Tablets Section No. 6 Group D

Devlopement and Evaluation of Simvastatin Nanoparticles using Nanosuspension Technique

International Journal of Pharma and Bio Sciences V1(1)2010 UV- SPECTROPHOTOMETRIC DETERMINATION OF TENATOPRAZOLE FROM ITS BULK AND TABLETS

The Nitrofurantoin Capsules Revision Bulletin supersedes the currently official monograph.

The Influence of Hydro-Alcoholic Media on Hypromellose Matrix Systems

Zero And First Order Derivative Spectrophotometric Methods For Determination Of Dronedarone In Pharmaceutical Formulation

In the present study, parameters such as solubility, medium ph, surfactant type and dissolution behavior of formulations,

Simultaneous UV Spectrophotometric Method for the Estimation of Cefuroxime Axetil and Probenecid from Solid Dosage Forms

Research Article. Solubility and dissolution rate enhancement of efavirenz by inclusion complexation and liquid anti-solvent precipitation technique

Journal of Chemical and Pharmaceutical Research

Enhancement of dissolution of Lercanidipine Hydrochloride using Solid Dispersion Technique

Design and development of fast dissolving tablets containing ziprasidone by solid dispersion method

Journal of Pharmaceutical and Scientific Innovation

Int. J. Pharm. Sci. Rev. Res., 30(2), January February 2015; Article No. 09, Pages: 63-68

Spectrophotometric Determination of Lorsartan Potassium and its Dosage Form by Bromothymol Blue and Phosphate Buffer

International Journal of Pharma and Bio Sciences SPECTROPHOTOMETRIC DETERMINATION OF DIACERIN IN BULK AND PHARMACEUTICAL FORMULATION

Plop Plop, Fizz Fizz, Oh What A Relief It Is (Which Pain Reliever Works Fastest)

Comparative Studies on Dissolution Enhancement of Glibenclamide in Solid Dispersions Made by Different Techniques

COMPUTER AIDED DRUG DESIGN (CADD) AND DEVELOPMENT METHODS

Formulation and in vitro evaluation of candesartan liquid solid compacts to enhance drug solubility

7. Stability indicating analytical method development and validation of Ramipril and Amlodipine in capsule dosage form by HPLC.

DISSOLUTION ENHANCEMENT OF CURCUMIN BY HYDROXYPROPYL-β-CYCLODEXTRIN COMPLEXATION

Research Article. Development of Controlled Release Tablets of Nisoldipine with Improved Pharmaceutical Properties

Indian Journal of Research in Pharmacy and Biotechnology

A theoretical approach to evaluate the release rate of. acetaminophen from erosive wax matrix dosage forms

Impact factor: 3.958/ICV: 4.10 ISSN:

INTERNATIONAL RESEARCH JOURNAL OF PHARMACY

Ketorolac tromethamine (KT)[1] is

DEVELOPMENT AND VALIDATION OF A SPECTROPHOTOMETRIC METHOD FOR DETERMINATION OF DRONEDARONE IN BULK DRUG AND PHARMACEUTICAL FORMULATION

Experiment 24. Chemical recycling of poly(ethylene) terephthalate (PET)

Received: ; Accepted:

INVESTIGATING THE POTENTIAL OF POLYETHYLENE GLYCOLS IN SOLUBILIZATION OF IMIDAZOLE DRUGS OF INTEREST OMAR H. EL-GARHY

Transcription:

Enhanced Dissolution Rate of Atorvastatin Calcium using Solid Dispersion with PEG 6000 by Dropping Method Lakshmi Narasaiah.V 1*, Kalyan Reddy.B 1, Kishore.K 2, Raj Kumar.M 1, Srinivasa Rao.P 1, Venkateswara Reddy.B 3 1 Department of pharmaceutics, Dr. Samuel George Institute of Pharmaceutical Sciences, Markapur-523316, A.P, India; 2 Department of pharmacy, Andhra University, Vizag-530001, A.P, India.; 3 St.Pauls College of pharmacy, Ranga Reddy-500074, A.P, India. Abstract: The solid dispersion was defined as the dispersion of one or more active ingredients in an inert carrier or matrix. The purpose of the study was to improve the physicochemical properties of Atorvastatin like solubility, dissolution properties and stability of poorly soluble drug by forming dispersion with Poly Ethylene Glycol (PEG) 6000 as water soluble carrier. Atorvastatin was formulated by physical mixtures and solid dispersions (dropping method) using 1:1, 1:2 and 1:3 ratios of drug and polymer (PEG-6000). The saturation solubility was carried using water and phosphate buffer solution (PBS) and showed marked increase in the saturation solubility than that of pure drug. Higher in vitro dissolution of solid dispersions was recorded compared to their corresponding physical mixtures and the pure drug. The dispersion with PEG-6000 (1:3) by dropping method showed faster dissolution rate (89.65%) as compared to other dispersions with PEG-6000 (1:1 and 1:2) whichever prepared by physical mixture and dropping method. The FT-IR shows the complexation and there were no interactions. Finally solid dispersion of Atorvastatin: PEG-6000 prepared as 1:3 ratio by dropping method showed excellent physicochemical characteristics and was found to be described by dissolution release kinetics and was selected as the best formulation in this study. Key words: Solid Dispersion, Dropping Method, Water Soluble Carrier, Atorvastatin calcium and PEG-6000. Introduction: Atorvastatin calcium is anti-hyper lipidemic drug that is poorly soluble in water, having a biological half-life of 14 hrs and is a selective, competitive inhibitor of HMG-CoA reductase, the rate-limiting enzyme that converts 3-hydroxy-3- methylglutaryl-coenzyme A to mevalonate, a precursor of sterols, including cholesterol. By inhibiting de novo cholesterol synthesis, they deplete the intracellular supply of cholesterol [1]. Polyethylene glycol (PEG) is used for the preparation of solid dispersions. A particular advantage of PEGs for the formation of solid dispersions is that they have good solubility in many organic solvents. The melting point of PEGs lies below 65 0 C in all cases [2], which is advantageous for the manufacture of solid dispersions. Additional attractive features of PEGs include their ability to solubilize some compounds [3] and also improve compound wettability. Up to 40 percent of new chemical entities discovered by the pharmaceutical industry today are poorly soluble or lipophilic compounds. The solubility issues complicating the delivery of these new drugs also affect the delivery of many existing drugs [4]. Poorly water-soluble drugs show unpredictable absorption, since their bioavailability depends upon dissolution in the gastrointestinal tract [5, 6, 7]. The dissolution characteristics of poorly soluble drugs can be enhanced by several methods [8, 9, 10]. Lipophilic molecules, especially those belonging to the biopharmaceutics classification system (BCS) class II and IV, dissolve slowly, poorly and irregularly, and hence pose serious delivery challenges, like incomplete release from the dosage form, poor bioavailability, increased food effect, and high inter-patient variability [11]. Alteration of the solid state at the particle or molecular level involves a physical change in the drug and is an attractive option for improving drug solubility [12]. Particle size reduction by micronization or nanonization can enhance the dissolution rate; however, the apparent solubility remains unaltered. At the molecular level, polymorphs offer a limited solubility advantage because of a small difference in free energy. In contrast, amorphous systems with excess thermodynamic properties and lower energetic barrier can 484

offer significant solubility benefits [13]. This solubility benefit can be further enhanced by preparing solid dispersions (SDs). SDs contributes by slowing devitrification, enhancing wettability and modulating the properties of the solvent [14]. Solid dispersions (SDs) with superior pharmaceutical properties can be formulated into suitable dosage forms especially for geriatric population which exhibits variable drug responses due to many age related physiological changes coupled with disease states. Geriatric patients with lower GI motility and gastric emptying, fluctuating gastric ph and reduced intestinal blood flow rate exhibit variable absorption upon administration of solid dosages forms. Solid dispersion is one of the effective and widely used techniques for dissolution enhancement [15]. The two basic procedures used to prepare solid dispersions are the melting or fusion [16] and solvent evaporation [17] techniques. Therefore, in the present study, PEG 6000 was chosen as a suitable polymer for the preparation of solid dispersions. Solid dispersions were then evaluated by dissolution and FT-IR spectroscopy. Materials and Methods: Materials Atorvastatin Calcium was a gift sample from Dr.Reddy s lab, Hyderabad, India. Poly ethylene glycol 6000 was purchased from Merk, Mumbai, Potassium dihydrogen orthophosphate (Qualigens fine chemicals, Mumbai, India), Sodium hydroxide (Finar chemicals ltd. Ahemdabad, India) and methanol (Research-Lab fine chemicals industries, Mumbai, India). All required chemicals were analytical grade. Methods Preparation of Physical Mixture: Physical mixtures of atorvastatin calcium at three different mass ratios (1:1, 1:2 and 1:3) were prepared in a glass mortar by light trituration for 5 minutes. The mixtures were passed through a sieve no: 60. The prepared mixtures were then filled in hard gelatin capsules, sealed and stored in a dessicator until further use. The composition of F1, F2 and F3 formulations was shown in Table 1. Preparation of Solid Dispersion by Dropping Method: For the preparation of the Atorvastatin Calcium (ATC) solid dispersion prepared by dropping method, containing different weight ratios of ATC in PEG 6000. The composition of S1, S2 and S3 formulations was shown in Table 1. The PEG was melted in a porcelain dish at 58 0 C (±1 0 C) and a measured amount of Atorvastatin Calcium were added and stirred. The melted drug carrier mixture was pipetted and placed into an adjustable heating device to keep the temperature constant. The melted drug carrier mixture was dropped onto a stainless steel plate, where it solidified into round particles. The temperature of the stainless steel plate was <20 0 C. The round particles (equivalent to 80 mg of ATC) were placed into hard gelatin capsules (size no. 2) for further investigations. Physicochemical Characterization: Solubility Measurements: Phase and saturation solubility studies were performed according to the method described by Higuchi and Connors [18]. The saturation solubility of drug, physical mixtures and SDs with PEG 6000 (1:1, 1:2 and 1:3 w/w) in distilled water and phosphate buffer (ph 6.8) was determined by adding an excess of drug, physical mixture and SDs to 50 ml distilled water or Phosphate buffer in conical flask and were rotated in a orbital shaking incubator for 96 hrs at 37 0 C ±0.5 0 C. The saturated solutions were filtered through a 0.45 μm membrane filter, suitably diluted with water, phosphate buffer and analyzed by Elico SL-150 UV spectrophotometer at 245nm. FT-IR Spectroscopy: Infrared (IR) spectroscopy was conducted using Thermo Nicolet Nexus 670 Spectrophotometer and the spectrum was recorded in the wavelength region of 6000 485

Table 1: Composition of Atorvastatin Calcium Physical Mixtures and Solid Dispersions Ingredients (mg) F1 F2 F3 S1 S2 S3 ATC 80 80 80 80 80 80 PEG-6000 80 160 240 - - - PEG-6000 - - - 80 160 240 ATC: Atorvastatin calcium; PEG-Poly Ethylene Glycol to 500 cm -1. The procedure consisted of dispersing a sample (drug alone or mixture of drug and excipients) in KBr and compressing into discs by applying a pressure. The pellet was placed in the light path and the spectrum was obtained. Drug content analysis: The drug content in each solid dispersion and physical mixture was determined by the UV-Spectroscopic method. An accurately weighed quantity of solid dispersion or physical mixture, equivalent to 80 mg of atorvastatin calcium, was transferred to a 100 ml volumetric flask containing 10 ml of methanol and dissolved. The volume was made up to 100 ml with ph 6.8. The solution was filtered and the absorbance was measured after suitable dilutions by using Elico SL- 150 UV-Spectrophotometer at 245nm [19]. In Vitro dissolution studies: Dissolution rate studies were performed in ph 6.8 phosphate buffer at 37 ± 0.5 0 C, using 8-station USP type-ii apparatus with paddle rotating at 50 rpm. Solid products, solid dispersions as well as physical mixtures, each containing 80 mg of drug were subjected to dissolution. At fixed time intervals, samples withdrawn were filtered and spectrophotometrically analyzed for the drug content at 245 nm. Each test was performed in triplicate (n=3). Dissolution efficiency (DE) was calculated from the area under the dissolution curve at time t (using the trapezoidal rule) [20]. The similarity factor (f2) was evaluated to compare ATC release profiles. n f2 = 50 log {[1+1/n ( Rt Tt) 2 ] - t 1 0.5 100} Where R t and T t were the cumulative percentage of drug released for reference and test assay at time t respectively, n was the number of time points. The FDA suggests that two dissolution profiles are declared to be similar if the value of f2 is between 50 and 100 [21]. Results and Discussion: The drug content in physical mixtures and solid dispersions was found to be in the range of 95.56 % to 99.6 %. Therefore, dropping method used in this study appears applicable for the preparation of solid dispersions without affecting drug content. Solubility Studies: The results of saturation solubility studies are given in Table 2. The solubility of pure drug in water and in PBS (ph 6.8) was found to be 27.04 ± 0.56 and 57.06 ± 0.67 μg/ml. The solubility of physical mixture prepared using PEG 6000 in the ratio (1:1, 1:2 and 1:3) was 35.59 ± 1.12, 46.87 ± 1.24, 57.79 ± 1.35 μg/ml in water and 63.78 ± 1.19, 72.76 ± 1.21, 81.89 ± 2.35 μg/ml in PBS. The solubility of SDs using PEG 6000 (1:1, 1:2 and 1:3) in water were found to be 38.52 ± 1.15, 47.83 ± 1.45, 59.56 ± 1.39 μg/ml and in PBS (ph 6.8) 67.96 ± 1.27, 74.67 ± 1.47 and 95.72 ± 1.48 respectively. 486

Table 2: Solubility studies and drug content of pure drug, physical mixtures and solid dispersions Formulation code Solubility (µg/ml) Water PBS Drug content (%) Pure drug 27.04 ± 0.56 57.06 ± 0.67 95.56±0.023 F-1 35.59 ± 1.12 63.78 ± 1.19 98.54±0.031 F-2 46.87 ± 1.24 72.76 ± 1.21 97.25±0.053 F-3 57.79 ± 1.35 81.89 ± 2.35 99.25±0.043 S-1 38.52 ± 1.15 67.96 ± 1.27 99.42±0.041 S-2 47.83 ± 1.45 74.67 ± 1.41 96.20±0.036 S-3 59.56 ± 1.39 95.72 ± 1.48 99.60±0.023 PBS: Phosphate Buffer solution Table 3: In vitro dissolution data for pure drug, marketed tablet, physical mixtures and solid dispersions Time (min) Pure drug Marketed tablet Cumulative % drug released F1 F2 F3 S1 S2 S3 10 15.633 21.84 19.102 20.958 23.161 20.044 21.870 24.073 20 20.175 25.807 22.469 26.455 27.674 24.243 26.028 28.739 30 23.197 30.728 26.705 31.191 32.112 28.484 30.715 32.970 40 26.898 34.935 31.455 36.105 36.378 32.300 34.877 49.303 50 30.883 50.477 35.914 52.431 57.332 36.668 53.108 64.958 60 35.261 63.052 50.701 66.778 70.421 63.139 68.644 75.635 70 51.006 77.222 64.010 75.351 82.393 78.784 84.631 89.652 All of the test samples showed an increase in drug solubility. As the solid dispersion is a metastable form and tends to transform in to the stable form, the drug concentration may tend to decrease with elapse of time during the solubility test. In order to avoid this problem all the solubility test samples of the different formulations were withdrawn and analyzed at established time (96hrs). This allowed readily comparing the solubility of different solid dispersions. 487

Table 4: Dissolution Kinetics of Atorvastatin Calcium by Physical Mixtures and Solid Dispersions Formulated With PEG-6000 Formulati on code Zero order Correlation Coefficient (R 2 ) First order Higuchi Peppas Hixson Crowell Slope (n) DE 30% DE 60% F1 0.9209 0.9176 0.9154 0.9052 0.9261 0.4756 19.60 26.95 F2 0.9543 0.9359 0.9235 0.9209 0.9556 0.5601 22.50 34.13 F3 0.9693 0.9123 0.9146 0.9029 0.9424 0.5508 23.91 36.30 S1 0.9320 0.8770 0.8744 0.8783 0.9043 0.5359 20.91 29.78 S2 0.9483 0.8886 0.8905 0.8832 0.9209 0.5496 22.68 33.67 S3 0.9714 0.9212 0.9250 0.9149 0.9553 0.5895 24.72 40.19 *DE 30 and DE 60, dissolution efficiency at 30 and 60 minutes. % Drug Released 90 80 70 60 50 40 30 20 10 0 0 10 20 30 40 50 60 70 80 Time (min) Pure Drug F1 F2 F3 Figure 1: Dissolution Profiles of Atorvastatin Calcium Physical Mixtures Formulated With PEG-6000. The solubility of different concentrations of drug and polymer was observed and the prepared formulation with PEG 6000, 1:3 (Drug: Carrier) presented higher dissolution concentration as compared with the other formulations obtained with different ratios (1:1 and 1:2). Maximum solubility in PBS was observed in dropping method 1:3 (Drug: PEG 6000) ratio 95.72 ± 1.48 μg/ml, when compared % Drug Released 100 90 80 70 60 50 40 30 20 10 0 0 10 20 30 40 50 60 70 80 Time (min) Pure Drug S1 S2 S3 Figure 2: Dissolution Profiles of Atorvastatin Calcium pure drug and Solid Dispersions. with that of pure Atorvastatin Calcium(57.06 ± 0.67 μg/ml). In Vitro drug release: The dissolution profiles of Atorvastatin Calcium for solid dispersion and physical mixture performed in 6.8 phosphate buffer were as shown in Table 3. The dissolution rate was significantly increased when the Atorvastatin Calcium: PEG 6000 ratio was at 1:3. 488

100 90 80 70 % Drug Released 60 50 40 30 20 10 0 0 10 20 30 40 50 60 70 80 Time (min) Pure Drug Marketed S3 Figure 3: Dissolution profile of Pure Atorvastatin Calcium, Marketed tablet and Solid dispersion (S3) The mean percentage of drugs for physical mixture after 70 minutes was 64.01%, 75.35%, 82.39% for 1:1, 1:2, 1:3 respectively as shown in Figure 1 and 51.01%, 77.22% for pure drug and marketed tablet. But in the dropping Figure 4: FTIR Spectra of Atorvastatin Calcium method half fold increase in release rate was observed as 78.78%, 84.63% and 89.65% respectively as shown Figure 2 (This may be due to impact of complexation and bond formation. 489

Figure 5: FTIR Spectra of Atorvastatin Calcium & PEG-6000 Mixture This may lead improved solubility by reducing particle size).the drug release from all the formulations followed zero order kinetics, as the plot observed in between amount of drug released Vs time was found to be linear. To analyze the mechanism of drug release from these formulations, the data were followed Hixson Crowell equation ({fraction unreleased} 1/3 Vs time). The release kinetics & dissolution efficiency at 30 & 60 minutes (DE 30 & DE 60 ) for these formulations were given in Table 4. The slope values (n) obtained to decline between 0.4756 to 0.5895 for all formulations for the release of ATC, indicating non-fickian diffusion. The dissolutions profile showed in (Figure 3) and similarity factor (f 2 ), these two formulations were found to be 80.58% indicating the significant differences in between the selected (S3) and marketed tablet (Lipitor). The drug release from different formulations followed the order: S3>S2>S1. The above results indicated that the increasing concentration of PEG- 6000 content enhanced the drug release. Spectroscopy studies: The IR spectra of pure ATC and solid dispersions are shown in Figures 4 & 5. The IR spectra of pure ATC showed characteristic peaks at 2955.15 cm -1 (C-H stretching), 1313.56 cm -1 (C-N stretching), 3059.15 cm-1 (C-HO - stretching alcoholic group), 1564.97 cm -1 (C=O stretching amidic group), 3403.27 cm -1 (N-H - stretching), 1656.97 cm -1 (C=C - bending), 751.62 cm -1, 696.95 cm - 1 (C-F- stretching), 1104.39 cm -1 (O-Hbending). It might be the possibility of intermolecular hydrogen bonding between adjunct ATC molecules. The spectrum of pure ATC was equivalent to the spectra obtained by the addition of carrier. This indicated that no interaction occurred with a solid dispersion of drug and lipid carriers. The results revealed no considerable changes in the IR peaks of ATC, when mixed with polymer PEG- 6000. These observations indicated the compatibility of PEG-6000 with ATC. Conclusion: The prepared solid dispersions were extended to various characterizations. FTIR shows there was no degradation of 490

drug. The solubility and dissolution studies showed there is a possibility of improved solubility of Atorvastatin Calcium through solid dispersion with Poly ethylene glycol 6000. A maximum increase in dissolution rate was obtained with Atorvastatin Calcium: PEG 6000 solid dispersion with a weight ratio of 1:3 (drug: carrier). PEG 6000 dispersion by dropping method showed faster dissolution rate when compared with that of pure drug and physical mixtures. Finally it could be concluded that solid dispersion by dropping method for Atorvastatin Calcium using hydrophilic polymers would improved the aqueous solubility, dissolution rate and thereby enhancing its systemic availability. Acknowledgement: Authors are thankful to Dr. A.Satish, Secretary of Dr. Samuel George Educational society, Markapur for the financial support and infrastructural facilities for the study. References: [1] N.Arunkumar, M.Deecaraman, C.Rani, K.P.Mohanraj, K.Venkates Kumar. International Journal of PharmTech Research. 2009; 1(4), 1725-1730. [2] J. C. Price. American Pharmaceutical Association, Washington, 1994; 355 361. [3] G. V. Betageri and K. R. Makarla. Int. J. Pharm., 1995; 126, 155 160. [4] M. Hite, S. Turner and C. Federici. 2003; www. scolr.com/lit/pmps_2003_1.pdf. [5] A. H. Goldberg, M. Gibaldi and J. L. Kanig. J. Pharm. Sci., 1965; 54, 1145-1148. [6] A. H. Goldberg, M. Gibaldi and J. L. Kanig. J. Pharm. Sci., 1966; 55, 482-487. [7] A. H. Goldberg, M. Gibaldi, J. L. Kanig and M. Mayersohn. J. Pharm. Sci., 1966; 55, 581-583. [8] D. Hoerter and J. B. Dressman. Adv. Drug Del. Res., 1997; 25, 3-14. [9] B. C. Hancock and G. Zografi. J. Pharm. Sci., 1997; 86, 1-12. [10] T. Loftsson and M. E. Brewster. J. Pharm. Sci., 1996; 85, 1017-1025. [11] R. Lobenberg and G. L. Amidon. Eur. J. Pharm. Biopharm., 2000; 50, 3-12. [12] L. F. Huang and W. Q. Tong. Adv. Drug Del. Rev., 2004, 56, 321-334. [13] M. Pudipeddi and A. T. M. Serajuddin. J. Pharm. Sci., 2005; 94, 929-939. [14] D. Q. M. Craig. Int. J. Pharm., 2002; 231, 131-144. [15] W. L. Chiou and S. Riegelman, Pharmaceutical applications of solid dispersions. J. Pharm. Sci., 1971; 60, 1281-1302. [16] K. Sekiguchi, N. Obi. Chem. Pharm. Bull., 1961; 9, 866-872. [17] T. Tachibana, A. Nakamura. Kolloid-Z. Polym., 1965; 203, 130-133. [18] T. Higuchi, K. Connors. Adv. Anal. Chem. Instrum., 1965; 4, 117-123. [19] N.Arunkumar, M.Deecaraman, C.Rani, K.P.Mohanraj, K.Venkates Kumar. International Journal of PharmTech Research, 2009; 1(4), 1725-1730. [20] Khan KA. J Pharm Pharmacol. 1975; 27(1), 48-9. [21] P. Vijayalakshmi, V. Kusum Devi. Drug Dev. Ind. Pharmacy, 2008; 34, 33-45. 491