Preparation, Characterization and Antiinflammatory Activity of Celecoxib-β-Cyclodextrin Inclusion Complexes

Similar documents
CHAPTER VI FACTORIAL STUDIES ON THE EFFECTS OF CYCLODEXTRINS AND SOLUTOL HS15 ON THE SOLUBILITY AND DISSOLUTION RATE OF EFAVIRENZ AND RITONAVIR

A FACTORIAL STUDY ON ENHANCEMENT OF SOLUBILITY AND DISSOLUTION RATE OF IBUPROFEN BY β CYCLODEXTRIN AND SOLUTOL HS15

ENHANCEMENT OF SOLUBILITY AND DISSOLUTION RATE OF NIMESULIDE BY CYCLODEXTRINS, POLOXAMER AND PVP

EFFECT OF PVP ON CYCLODEXTRIN COMPLEXATION OF EFAVIRENZ FOR ENHANCING ITS SOLUBILITY AND DISSOLUTION RATE

FACTORIAL STUDIES ON THE EFFECTS OF HYDROXY PROPYL β- CYCLODEXTRIN AND POLOXAMER 407 ON THE SOLUBILITY AND DISSOLUTION RATE OF BCS CLASS II DRUGS

International Journal of Pharma and Bio Sciences

PREPARATION, CHARACTERIZATON AND DISSOLUTION KINETICS OF CELECOXIB : β AND HP β -CYCLODEXTRIN COMPLEXES

Comparative study of different solubility enhancement techniques on dissolution rate of zaltoprofen

Chemate and Chowdary, IJPSR, 2012; Vol. 3(7): ISSN:

FORMULATION AND CHARACTERIZATION OF TELMISATAN SOLID DISPERSIONS

3.1 Background. Preformulation Studies

SOLID INCLUSION COMPLEXES OF CLASS II IMIDAZOLE DERIVATIVE WITH Β CYCLODEXTRIN. Pradesh.

Mixed Hydrotropy: Novel Science of Solubility Enhancement

A STUDY ON SUITABILITY OF NIMESULIDE-BETACYCLODEXTRIN COMPLEX IN ORAL AND TOPICAL DOSAGE FORMS

International Journal of Pharma Sciences and Scientific Research

IJPAR Vol.3 Issue 4 Oct-Dec-2014 Journal Home page:

MEDAK DIST. ANDHRA PRADESH STATE, INDIA. Research Article RECEIVED ON ACCEPTED ON

Development, Estimation and Validation of Lisinopril in Bulk and its Pharmaceutical Formulation by HPLC Method

FORMULATION AND EVALUATION OF VALSARTAN TABLETS EMPLOYING CYCLODEXTRIN-POLOXAMER 407-PVP K30 INCLUSION COMPLEXES

ENHANCEMENT OF SOLUBILITY, DISSOLUTION RATE AND BIOAVAILABILITY OF RITONAVIR BY CYCLODEXTRINS AND SOLUTOL HS15 - A FACTORIAL STUDY

ENHANCEMENT OF SOLUBILITY OF BICALUTAMIDE DRUG USING SOLID DISPERSION TECHNIQUE

Journal of Pharmaceutical and Scientific Innovation

DRUG-EXCIPIENTS INTERACTION AND SOLUBILITY ENHANCEMENT STUDY OF SIMVASTATIN

7. SUMMARY, CONCLUSION AND RECOMMENDATIONS

RP-HPLC Analysis of Temozolomide in Pharmaceutical Dosage Forms

ISSN: ; CODEN ECJHAO E-Journal of Chemistry 2011, 8(3),

Asian Journal of Pharmacy and Life Science ISSN Vol. 1 (4), Oct-Dec, 2011

ARTESUNATE TABLETS: Final text for revision of The International Pharmacopoeia (December 2009) ARTESUNATI COMPRESSI ARTESUNATE TABLETS

Reverse Phase HPLC Analysis of Atomoxetine in Pharmaceutical Dosage Forms

A FACTORIAL STUDY ON THE ENHANCEMENT OF DISSOLUTION RATE OF KETOPROFEN BY SOLID DISPERSION IN COMBINED CARRIERS

Available online at

Preparation and Evaluation of Ethyl Cellulose Coated Microcapsules of Carbamazepine for Controlled Release

Tentu Nageswara Rao et al. / Int. Res J Pharm. App Sci., 2012; 2(4): 35-40

Development and validation of stability indicating RP-LC method for estimation of calcium dobesilate in pharmaceutical formulations


Scholars Research Library. Der Pharmacia Lettre, 2016, 8 (6): (

International Journal of Research in Pharmaceutical and Nano Sciences Journal homepage:

The Nitrofurantoin Capsules Revision Bulletin supersedes the currently official monograph.

Inclusion Complexes of Ketoprofen with β-cyclodextrins: Oral Pharmacokinetics of Ketoprofen in Human

ISSN (Print)

Comparative Dissolution Study of Glipizide by Solid Dispersion Technique

A HIGH PERFORMANCE LIQUID CHROMATOGRAPHIC ASSAY FOR LERCANIDIPINE HYDROCHLORIDE

World Journal of Pharmaceutical Research

Lipid Based Matrices as Colonic Drug Delivery System for Diflunisal (In-vitro, In-vivo study)

DEVELOPMENT AND VALIDATION OF RP-HPLC METHOD FOR QUANTITATIVE ANALYSIS TOLBUTAMIDE IN PURE AND PHARMACEUTICAL FORMULATIONS

Int. Res J Pharm. App Sci., 2013; 3(6):42-46 ISSN:

Development and Validation for Simultaneous Estimation of Sitagliptin and Metformin in Pharmaceutical Dosage Form using RP-HPLC Method

FORMULATION AND EVALUATION OF ETORICOXIB TABLETS EMPLOYING CYCLODEXTRIN- POLOXAMER PVPK30 INCLUSION COMPLEXES

Bioequivalence Studies of Two Formulations of Famciclovir Tablets by HPLC Method

skim milk as carrier by kneading method. They were evaluated for percentage yield, drug content, FT-IR

Preparation of 200 mg fenofibrate hard capsule with high dissolution profile with microparticle entrapped micelles technology

Pelagia Research Library

Umapathi et al. Keywords: Fixed dose combination; Rifampin quinone; Tuberculosis; Sodium ascorbate; Quantitative determination; HPLC

DEVELOPMENT AND VALIDATION OF NEW HPLC METHOD FOR THE ESTIMATION OF PALIPERIDONE IN PHARMACEUTICAL DOSAGE FORMS

Development of a Validated RP-HPLC Method for the Analysis of Citicoline Sodium in Pharmaceutical Dosage Form using Internal Standard Method

Jagua (Genipin-Glycine) Blue (Tentative)

Abstract. G. D. Patil 1, P. G. Yeole 1, Manisha Puranik 1 and S. J. Wadher 1 *

RP-HPLC Method Development and Validation of Abacavir Sulphate in Bulk and Tablet Dosage Form

HPLC-UV Determination of Abacavir Sulphate in Pharmaceutical Dosage Forms

DEVELOPMENT OF RP-HPLC METHOD FOR ESTIMATION OF DROTAVERINE HYDROCHLORIDE IN PHARMACEUTICAL FORMULATIONS

Development and Validation of a New Uv Method for the Analysis of Rebamipide

RP- HPLC and Visible Spectrophotometric methods for the Estimation of Meropenem in Pure and Pharmaceutical Formulations

RITONAVIRI COMPRESSI RITONAVIR TABLETS. Final text for addition to The International Pharmacopoeia (July 2012)

Development and validation of spectrophotometric method for simultaneous estimation of Sumatriptan and Naproxen sodium in tablet dosage form


Available online at

Determination of 6-Chloropicolinic Acid (6-CPA) in Crops by Liquid Chromatography with Tandem Mass Spectrometry Detection. EPL-BAS Method No.

Isocratic Reversed Phase Liquid Chromatographic Method Validation for the Determination of Cilostazol in Pure and Formulations

TENOFOVIR TABLETS: Final text for addition to The International Pharmacopoeia (June 2010)

Formulation Development of Aceclofenac Tablets Employing Starch Phosphate -A New Modified Starch

Asian Journal of Pharmaceutical Analysis and Medicinal Chemistry Journal home page:

EVALUATION OF EFFERVESCENT FLOATING TABLETS. 6.7 Mathematical model fitting of obtained drug release data

Fluorescent Carbon Dots as Off-On Nanosensor for Ascorbic Acid

DRAFT PROPOSAL FOR THE INTERNATIONAL PHARMACOPOEIA: CARBAMAZEPINI COMPRESSI - CARBAMAZEPINE TABLETS

RP-HPLC METHOD DEVELOPMENT AND VALIDATION FOR THE ESTIMATION OF BACLOFEN IN BULK AND PHARMACEUTICAL DOSAGE FORMS

Rebaudioside a From Multiple Gene Donors Expressed in Yarrowia Lipolytica

INTERNATIONAL JOURNAL OF INSTITUTIONAL PHARMACY AND LIFE SCIENCES

PHARMA SCIENCE MONITOR AN INTERNATIONAL JOURNAL OF PHARMACEUTICAL SCIENCES

Preparation and Physicochemical Properties of the Complex of Naringenin with Hydroxypropyl-β-Cyclodextrin

A Comparative Evaluation of Cross Linked Starch Urea-A New Polymer and Other Known Polymers for Controlled Release of Diclofenac

Journal of Chemical and Pharmaceutical Research, 2018, 10(3): Research Article

F. Al-Rimawi* Faculty of Science and Technology, Al-Quds University, P.O. Box 20002, East Jerusalem. Abstract

Comparison of conventional HPLC with UPLC method for determination of albuterol sulfate and it s impurities in pharmaceutical formulation

Adsorption and Dehydration of Water Molecules from α, β and γ Cyclodextrins-A study by TGA analysis and gravimetry

KEY WORDS Chitosan, Oats powder, Solid dispersion, Simvastatin, Oral bioavailability, Karaya Gum.

DESIGN AND EVALUATION OF CONTROLLED RELEASE MATRIX TABLETS OF FLURBIPROFEN

The Effect of Water Soluble Polymers on Felodipine Aqueous Solubility and Complexing Abilities with Natural and Modified β-cyclodextrin

A FACTORIAL STUDY ON THE ENHANCEMENT OF DISSOLUTION RATE OF ACECLOFENAC BY SOLID DISPERSION IN STARCH PHOSPHATE AND GELUCIRE

Influence of External Coagulant Water Types on the Performances of PES Ultrafiltration Membranes

Preparation and Characterization of Candesartan Cilexetil Solid Lipid Nanoparticulate Capsules

FORMULATION AND EVALUATION OF MELT-IN-MOUTH TABLETS OF DOMPERIDONE CONTAINING MULTICOMPONENT INCLUSION COMPLEX

A study on the effects of different surfactants on Ethylcellulose microspheres

Simultaneous estimation of Metformin HCl and Sitagliptin in drug substance and drug products by RP-HPLC method

Residue Monograph prepared by the meeting of the Joint FAO/WHO Expert Committee on Food Additives (JECFA), 82 nd meeting 2016.

Characterization and Solubility Studies of Mefloquinehydrochloride Inclusion Complexes with α - Cyclodextrin/ Hydroxypropyl α -Cyclodextrin.

SIMULTANEOUS DETERMINATION OF ATORVASTATIN AND EZETIMIBE BY RP-HPLC IN PURE AND PHARMACEUTICAL DOSAGE FORM

SIMULTANEOUS ESTIMATION OF VALSARTAN AND HYDROCHLOROTHIAZIDE IN TABLETS BY RP-HPLC METHOD

Lutein Esters from Tagetes Erecta

Pharmaceutical Studies on Formulation and Evaluation of Sustained Release Tablets Containing Certain Drugs

LEVONORGESTREL AND ETHINYLESTRADIOL TABLETS. (January 2012) DRAFT FOR COMMENT

Transcription:

Asian Journal of Chemistry Vol. 21, No. 3 (2009), 1759-1768 Preparation, Characterization and Antiinflammatory Activity of Celecoxib-β-Cyclodextrin Inclusion Complexes DEMET SENSY*, ÜMIT GÖNÜLLÜ, LCAY SAGIRLI, GÜLGÜN YENER and TUNCAY ALTUG Department of Pharmaceutical Technology, Faculty of Pharmacy Istanbul University, Universite-34116, Istanbul, Turkey Fax: (90)(212)4400260; Tel: (90)(212)4400000 E-mail: demetsensoy@istanbul.edu.tr The influence of β-cyclodextrin on the in vitro dissolution rate, in vivo absorption and oral bioavailability of a poorly water soluble antiinflammatory agent, celecoxib was studied. For this purpose, celecoxib and β-cyclodextrin complexes were prepared in 1:1 and 1:2 molar ratios by the physical mixture, kneading and freeze-drying methods. The complexes were preliminary confirmed using differential scanning calorimetry, fourier transform-infrared spectroscopy and scanning electron microscopy. The solubility studies revealed a linear relationship between the increase in celecoxib solubility and the increase in β-cyclodextrin concentration. The in vitro dissolution studies that were performed in phosphate buffer (ph 7.4) showed that celecoxib:β-cyclodextrin (1:2) solid complexes prepared by freeze-drying method had highest celecoxib release compared to the other solid complexes. Pharmacological studies were performed with this complex in a carrageenan induced rat hind paw oedema model. Regarding to the inhibition of edema (%) and swelling (%) results, celecoxib:β-cyclodextrin (1:2) binary mixture prepared by freeze-drying method showed significant improvement compared to pure drug. Key Words: Celecoxib, β-cyclodextrin, Solubility, Inclusion complexes, Antiinflammatory activity. INTRDUCTIN Celecoxib (CEL) 4-[5-(4-methylphenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl] benzenesulphonamide, a cyclooxygenase-2 (CX-2) (Fig. 1) selective inhibitor is a non-steroid antiinflammatory drug (NSAID) approved for the treatment of rheumatic pain and osteoarthritis 1,2. Most of NSADs inhibit both isoforms of cyclooxygenase (CX-1 and CX-2) and inhibit platelet aggregation. Because of CEL's specific CX-2 inhibition, it has less potential to cause gastropathy and risk of GI bleeding 3,4. Celecoxib is a poor water soluble drug and it has a pka 11.1 with low solubility Department of Analytical Chemistry, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Turkey. Experimental Animal-Research and Breeding Laboratory, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey.

1760 Sensoy et al. Asian J. Chem. (ca. 5 µg/ml). This fact contributes to high variability in absorption after oral administration and affects bioavailability. Thus, it is important to enhance the solubility and dissolution rate of CEL to improve its overall oral bioavailability 5. There have been many studies recorded related to the enhancement of CEL solubility in water by using cyclodextrins 6-8. H 3 N S N N CF 3 H 3 C Fig. 1. Chemical structure of celecoxib (CEL) Cyclodextrins are cyclic oligosaccharides composed of α-d-glucopyranose units joined through a 1-4 bond 9,10. During the past decades, cyclodextrins and their derivatives such as β-cyclodextrin (βcyd) (Fig. 2), α-cyclodextrin and γ-cyclodextrin have aroused considerable interest in the drug formulation because of their potential to form complexes with many water insoluble drug molecules 11,12. RH 2 C RH 2 C RH 2 C R R R R R R R R CH 2 R R R R R R R CH 2 R CH 2 R CH 2 R Fig. 2. Structural formula of β-cyclodextrin (βcyd) They are capable of forming water-soluble inclusion complexes with poorly water-soluble drugs by taking up a whole drug molecule or a part of it, into the cavity of the cyclodextrin molecule 13-15. Thus, they are commonly used in drug

Vol. 21, No. 3 (2009) Celecoxib-β-Cyclodextrin Inclusion Complexes 1761 formulations to enhance solubility, dissolution rate, stability and bioavailability by means of the formation of inclusion complexes 16-19. The purpose of this study is to improve the solubility and in vitro dissolution rate, in vivo absorption and oral bioavailability of CEL. Drug and βcyd solid complexes were prepared in 1:1 and 1:2 molar ratios by the three different methods: physical mixture, kneading and freeze-drying. Complex formations were confirmed using differential scanning calorimetry, fourier transform-infrared spectroscopy and scanning electron microscopy. Dissolution studies were performed for binary complex systems and the overall results were compared with those of pure CEL and physical mixtures. Furthermore, based on the in vitro evaluation of complexes, the most suitable binary complex chosen for pharmacological studies and bulk drug was compared in the model of carrageenan induced hind paw edema in the rat. EXPERIMENTAL Celecoxib (CEL) was supplied by Pfizer (Turkey), β-cyclodextrin (βcyd) was purchased from Cerestar (USA). ther reagents were of analytical quality. Phase solubility studies: Solubility study was carried out according to the method described by Higuchi and Connors 20. To 10 ml distilled water containing various concentrations of βcyd (from 0 to 0.012 M) excess amount of CEL (30 mg) were added and sonicated in an ultrasonic bath for 20 min and shaken in water bath at 37 ± 0.5 ºC until equilibrium was reached after ca. 48 h. The suspensions were filtered through 0.45 µm Millipore membrane filter. CEL concentration was determined by HPLC 16. The phase solubility was therefore obtained. The apparent 1:1 stability constant, Ks, was calculated from phase solubility diagrams using the equation 20 : Slope Ks = So(1 Slope) where So is CEL solubility in absence of βcyd (intercept). Preparation of inclusion complexes in solid state: Based on the results of the phase solubility studies, solid systems were prepared with 1:1 and 1:2 molar ratios of CEL and βcyd, using kneading and freeze-drying methods. Physical mixtures were prepared as a reference for the purpose of comparison. Physical binary mixture (PM): PMs were prepared by homogeneous blending (for 10 min) of previously sieved (60-160 µm) and weighed CEL and βcyd in a cubic mixer. Kneading binary system (KNE): βcyd was wetted in a ceramic mortar with distilled water until a paste obtained. The required amount of CEL was then slowly added and the slurry was kneaded for about 1 h. Further, the product was dried at 37 ± 0.5 C for 24 h and sieved through 125-500 µm sieve granulometric fraction. Lyophilized binary system (LPh): CEL and βcyd were added to basic aqueous solution, according to 1:1 and 1:2 (CEL:βCyD) molar ratios. The solution stirring was maintained for 48 h. Furthermore, the resultant solution was filtered through

1762 Sensoy et al. Asian J. Chem. 0.45 µm Millipore membrane filter, frozen and lyophilized in a freeze dryer (Lyovac GT-2, Leybold-Heraeus). Characterization of samples Thermal analysis by differential scanning calorimetry (DSC) was carried out using a Du Pont Instruments 910 apparatus. Samples (3 mg) of CEL, βcyd and binary systems were put into aluminium pans and scanned at the heating rate of 10 C min -1 over the temperature range of 30-250 C. FTIR spectrum of the CEL, βcyd, PMs, KNEs and LPhs were measured in potassium bromide disks using a Perkin- Elmer Model 1600 FTIR spectrometer (USA). SEM analysis of the CEL, βcyd and solid complexes were carried out using scanning electron microscope (Joel, JSM-5200, Japan) with an accelerating voltage of 20 kv and high vacuum operation. The samples were coated with gold-palladium mixture under an argon atmosphere at room temperature yielding a film thickness of 5 nm. In this study, an HPLC method was developed to determine drug concentration released from CEL-βCyD solid complexes. For this purpose, method reported by Schonberger et al. 21 was applied after some modifications. The Shimadzu (Kyoto, Japan) LC 10A liquid chromatography system used in the analysis was consisted of a model LC 10 AT solvent delivery system, a Rheodyne injection system with a loop of 20 µl and a model CT 10A column oven at 25 C. The components were detected at 390 nm emission wavelength (252 nm excitation) by using model RF 10 AXL spectrofluori-metric detector. Separation was performed on analytical (250 mm 4.6 mm) column with a guard column (3 mm 4.6 mm) packed with octadecyl (particle size 5 µm), phenomenex. The mobile phase was acetonitrile-water (70:30) at a flow rate 1.2 ml/min. Modified method was applied without internal standard to obtain the calibration equation. Calibration curve was linear over a range of 0.2-10 µg/ml. For the determination of drug concentrations in samples, filtrated samples were diluted with ultra purified water and 20 µl portions were injected into the HPLC system. The drug concentrations were determined by peak areas using the standard curve established from a series of standards with known concentrations. Under these conditions, the polymer did not interfere with the drug peak. In vitro dissolution studies: The dissolution profiles of pure drug and binary systems were carried out in 900 ml phosphate buffer (ph 7.4), using Aymes DT-6E model dissolution apparatus, according to the USP rotating paddle method. Powdered samples containing 20 mg CEL were used. The paddle speed was set at 100 rpm and the temperature was maintained at 37 ± 0.5 C. An aliquot of sample was withdrawn at specified time intervals, filtered through 0.45 µ nylon disc filter and analyzed by HPLC. Each test was carried out 3 times. Pharmacological studies: The antiinflammatory activity tests were performed in order to determine the possible enhancement in pharmacological activity of CEL after incorporation into inclusion form. For this purpose, the animals were housed in plastic cages at constant temperature (22 ± 1 C) and humidity (60 ± 1 %) under a 12 h light-dark cycle. They were given standard laboratory diet and tap water

Vol. 21, No. 3 (2009) Celecoxib-β-Cyclodextrin Inclusion Complexes 1763 ad libitum. The in vivo experimental protocol was approved by the Ethical Scientific Committee of Istanbul University, Cerrahpasa Medical Faculty and the experiments were carried out under veterinary supervision. The antiinflammatory study was performed using carrageenan induced rat hind paw edema method on Wistar Albino rats (150-200 g) 22-24. The animals were divided into 3 groups of 6 animals each. CEL:βCyD (1:2) LPh (equivalent to 20 mg/kg CEL) (Group 1) and pure drug (20 mg/kg) (Group 2) were administered orally as aqueous suspensions with 0.25 % sodium carboxymethyl cellulose. An aqueous solution of 0.25 % CMC was administered to control group (Group 3). After 0.5 h of oral irrigation of drugs, edema was induced by injecting 0.1 ml carrageenan (1 % w/v) into the plantar tissue of the hind-paw. The volume of the treated paw was measured with a plethysmometer at the scheduled time intervals for a period of 6 h and the swelling (%) and percentage inhibition of edema (%) were calculated with eqns. 1 and 2, respectively 25. Vt Vo Swelling (%) = 100 (1) Vo (Vt Vo) control (Vt Vo) treat Percentage inhibition of edema (%) = 100 (Vt Vo) (2) control Vo = Average volume in the hind paws of rats before any treatment, Vt = Average volume after inflammatory agent injection. RESULTS AND DISCUSSIN Phase solubility studies: The phase solubility diagram for the solid complex formation between CEL and βcyd is presented in Fig. 3. The graphical representation reveals a linear relationship (R 2 > 0.99) between the increase in CEL solubility and increase in βcyd concentration. The concentration of CEL in distilled water at ph 6.8, 25 C is clearly affected by the presence of βcyd. According to the Higuchi and Connors classification, the diagrams obtained were AL type 20. The apparent stability constant, Ks, obtained from the slope of the linear phase solubility diagram was found to be 1060.7 M-1, which indicates that, the CEL:βCyD complexes are adequately stable. 0.025 CEL concentration (mm) 0.020 0.015 0.010 0.005 y = 0.0018x + 0.0017 2 R = 0.9991 0.000 0 2 4 6 8 10 12 14 Fig. 3. Phase solubility diagram of CEL:βCyD in aqueous media at 37 ± 0.5 C (n = 3)

1764 Sensoy et al. Asian J. Chem. Thermal analysis: Thermograms regarding to DSC analysis performed on CEL, βcyd and binary systems. The thermogram of CEL presents an endothermic peak at 164.2 ºC which represents the melting point of the drug and βcyd presents an endothermic peak at 119.4 ºC. Peaks related to CEL were observed to be reduced both in physical binary mixture and kneading binary mixture. More reduction of CEL peaks were also seen in CEL:βCyD (1:2) KNE formulations compared to 1:1. The peak reduction may be explained by small CEL content but its displacement is supposed to be caused by an interaction with the CD during heating in case of inclusion complexes. The thermograms of CEL:βCyD (1:1) lyophilized binary system (LPh) displayed nearly no peak corresponding to CEL melting whereas there was absolutely no peak in case of CEL:βCyD (1:2) LPh. This situation may be explained as the complete complex formation between CEL an βcyd when used in CEL:βCyD (1:2) LPh formulations. Fourier transform-infrared spectroscopy (FTIR): FTIR spectra results of CEL, βcyd and binary systems were shown in Fig. 4. Characteristic bands of CEL were observed at 1596 cm -1 (-NH vibration), 1347 and 1134 cm -1 (S 2 asymmetric and symmetric tension), 3099 cm -1 (C-H stretching band), βcyd spectrum presents a large band and peak in the region of 3900-2900 cm -1, a shorter band between 1600 and 1700 cm -1 and a large band which displays distinct peaks, in the region of 1200-900 cm -1. When the spectra of the PMs, KNEs and LPhs were analyzed, it can be easily seen that peaks related to -NH vibration at 1596 cm -1 were disappeared in all formulations tested. This could be due to the complex formation of CEL to Fig. 4. FTIR spectras of CEL, βcyd and binary systems

Vol. 21, No. 3 (2009) Celecoxib-β-Cyclodextrin Inclusion Complexes 1765 βcyd from NH groups. In all complexes, C--R vibration band at 1638 cm -1 were seen due to βcyd and these bands disappeared in CEL. Strength of peaks in CEL:βCyD (1:2) LPh were lower than the one in 1:1. As expected, the lowest peak strengths were obtained in LPhs compared to KNEs and PMs. When peaks strength of CEL were analyzed, it could be seen that the highest peaks were observed which means that the lowering of CEL peaks strength show the complex formation between CEL and βcyd. Evaluation of DSC and FTIR analysis suggest stronger interaction between drug and CD in the solid complexes rather than in a simple physical binary mixture, as a result of CEL:βCyD complexation. Scanning electron microscopy (SEM): SEM is used to determine the microscopic properties of the CEL, βcyd and binary systems (Fig. 5). Therefore existence of the principal components in the complex and the possible enhancement effects of drying process on morphology of particles and solubility were analyzed. According to results obtained from SEM, pure CEL is characterized by the presence of a crystalline particle of regular size. Pure βcyd appears as crystalline particles without a definite shape. Physical binary mixtures, the characteristic CEL crystals, which were mixed with excipient particles or adhered to their surface, were clearly detectable, thus confirming the presence of crystalline drug. This fact was observed in physical mixtures between βcyd and other poorly water soluble drugs 26. In case of KNE products, it was possible to differentiate CEL crystals gathered on the surface of βcyd particles that had lost their original shapes and crystal sizes were smaller. It was observed that LPh products appeared to show lesser crystalline structure Fig. 5. SEM micrographs of CEL (a), βcyd (b), physical mixture (c), kneaded (d) and lyophilized (e) systems

1766 Sensoy et al. Asian J. Chem. with a soft and feathery appearance and crystals of the single components were still not detectable 27. Lyophilized binary mixture seems to have acquired the same shape, presenting some sticks covering its surface, with a shape similar to that of the drug (Fig. 5). Dissolution studies: The dissolution profiles of CEL and binary systems are given Fig. 6. CEL content in the solid complex formulations was assayed by a HPLC method which was developed to determine CEL content. The results of in vitro dissolution studies, CEL:βCyD (1:2) LPh showed the faster and highest CEL release (26.02 %) compared to the other formulations followed by CEL:βCyD (1:1) LPh (10.31 %), CEL:βCyD (1:2) KNE (6.11 %), CEL:βCyD (1:1) KNE (2.78 %), CEL:βCyD (1:2) PM (2.5 %) and CEL:βCyD (1:1) PM 2.03 %) at 15 min and 28.65, 13.42, 9.16, 5.53, 5.14, 3.19 % at 1 h, respectively. These results revealed that, released pure drug was 0.97 % at 15 min and 1.84 % at 1 h. Therefore solubility of CEL was enhanced by its complexation with βcyd as expected. The incorporation of CEL to βcyd in especially LPhs influenced its dissolution profile. Fig. 6. In vitro dissolution profiles of CEL and binary systems (n = 3) Per cent dissolved drug amount obtained with CEL:βCyD (1:2) LPh was found to give the best results especially when compared to PMs and pure drug, the amount dissolved was clearly high. This increase on CEL release is related to an improvement in its solubility caused by CEL:βCyD water-soluble inclusion due to preparation method and CEL:βCyD molar ratio. When the two preparation methods were compared inclusion complex prepared with freeze-drying method, in both ratios, released the drug faster than the complex prepared with kneading method. It was observed that the complexes with 1:1 ratio released less drug. However release rates of CEL were found very low both in physical binary mixtures and

Vol. 21, No. 3 (2009) Celecoxib-β-Cyclodextrin Inclusion Complexes 1767 which could be due to the incomplete complex formations. Similarly, CEL:βCyD (1:1) KNE formulation released drug very slowly as in CEL:βCyD (1:2) PM and not found suitable for use. It is possible to reduce surface tension between soluble drug and dissolution medium. By using hydrophilic cyclodextrin and enhancement of drug's wettability and formation of easily soluble complex may improve the dissolution rate gained by physical binary mixture. After preliminary studies, CEL:βCyD (1:2) LPh was found to be the most suitable one and chosen for in vivo experiments. Pharmacological studies: Antiinflammatory activity tests were performed to compare the CEL's affectivity alone and inside inclusion complex. Results of these tests were shown in Table-1. The percentage inhibition gained by CEL:βCyD (1:2) LPh which was shown to have fastest and highest active substances solubility by in vitro solubility studies is more than 50 % in 1 h. It reaches the maximum value (66.85 %) at 2 h. TABLE-1 RESULTS F PHARMACLGICAL STUDIES Time (h) 1 h 2 h 4 h 6 h Control 18.10±3.09 32.66±2.51 40.45±7.65 48.38±13.04 Pure CEL 16.41±4.03(15.28) 29.96±7.28(20.23) 34.74±13.25(20.57) 40.12±11.58(16.69) CEL:βCyD(1:2)LPh 8.48±1.16(53.08) 10.62±1.86(66.85) 16.92±5.09(55.50) 22.04±1.92(45.54) Data as presented as means ± SD, n = 6, of swelling (%) calculated from eqn. 1, with percentage inhibition of edema (%) calculated from eqn. 2 given in parenthesis. The swelling (%) were calculated 22.04, 40.12 and 48.38 % at 6 h for the CEL, CEL:βCyD (1:2) LPh and control group, respectively. When the swelling (%) and inhibition of edema (%) were analyzed, it can be easily seen that CEL:βCyD (1:2) LPh more effective than CEL for prevent of edema. However, both of therapy groups were achieved significant higher inhibition of the inflammation induced by carrageenan compared to control group (p < 0.05). This fact may be attributed to the capability of forming water-soluble inclusion complexes with poorly water-soluble drugs by taking up the whole drug into its cavity in case of CEL. This mechanism improves the solubility and therefore CEL's poor water solubility changes and CEL dissolves more rapidly and in high amounts when complexed with βcyd. This result is in accordance with results obtained from solubility tests as expected. Conclusion According to these findings, it could be concluded that this is an important attempt for modulation of CEL release and further an improvement is expected on the bioavailability. Due to all the results obtained, it could be concluded that CEL solubility and dissolution characteristics might be modulated by using βcyd inclusion complexes. Additionally, in vivo test results demonstrated that antiinflammatory activity of CEL:βCyD (1:2) LPh inclusion complex was found to be the most suitable.

1768 Sensoy et al. Asian J. Chem. REFERENCES 1. N.E. Lane, J. Rheumatol., 24, 20 (1997). 2. D. Clemett and K.L. Goa, Drugs, 59, 957 (2000). 3. H. Thakkar, R.K. Sharma, A.K. Mishra, K. Chuttani and R.R. Murthy, AAPS. Pharm. Sci. Tech., 6, Article 12 (2005). 4. M.M. Goldenberg, Clin. Ther., 21, 1497 (1999). 5. S. Paulson, M. Vaughn, S. Jessen, Y. Lawal, C. Gresk, B. Yan, T. Maziasz, C. Cook and A. Karim, J. Pharmacol. Exp. Ther., 297, 638 (2001). 6. K.P. Chowdary and S.V. Srinivas, AAPS. Pharm. Sci. Tech., 7, Article 79 (2006). 7. C.A. Ventura, I. Giannone, D. Paolino, V. Pistarà, A. Corsaro and G. Puglisi, Eur. J. Med. Chem., 40, 624 (2005). 8. M.S. Nagarsenker and M.S. Joshi, Drug Dev. Ind. Pharm., 31, 169 (2005). 9. J. Szejtli, Chem. Rev., 98, 1743 (1998). 10. R.A. Rajewski and V.J. Stella, J. Pharm. Sci., 85, 1142 (1996). 11. D.. Thompson, Crit. Rev. Ther. Drug Carrier Syst., 14, 1 (1997). 12. K. Uekama, F. Hirayama and T. Irie, Chem. Rev., 98, 2045 (1998). 13. A.P. Mukne and M.S. Nagarsenker, AAPS Pharm. Sci. Tech., 5, Article 19 (2004). 14. E.M. Martin Del Valle, Process. Biochem., 39, 1033 (2004). 15. K. Rajendrakumar, S. Madhusudan and T. Pralhad, Eur. J. Pharm. Biopharm., 60, 39 (2005). 16. T. Loftson, T.K. Guomundsdóttir and H. Fridriksdóttir, Drug Dev. Ind. Pharm., 22, 401 (1996). 17. T. Loftson and M.E. Brewster, J. Pharm. Sci., 85, 1017 (1996). 18. D. Duchene and D. Wouessidjewe, Drug Dev. Ind. Pharm., 16, 2487 (1990) 19.. Bekers, E.V. Uijtendal, J.H. Beijnen, A. Bult and W.J. Underberg, Drug Dev. Ind. Pharm., 17, 1503 (1991). 20. T. Higuchi and K.A. Connors, Adv. Anal. Chem. Instr., 4, 117 (1965). 21. F. Schönberger, G. Heinkele, T.E. Murdter, S. Brenner, U. Klotz and U. Hofmann, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci., 768, 255 (2002). 22. S. Baboota, M. Dhaliwal and K. Kohli, AAPS Pharm. Sci. Tech., 6, Article 83 (2005). 23. C.A. Winter, E.A. Risley and G.W. Nuss, Proc. Soc. Exp. Biol. Med., 111, 544 (1962). 24. W.L. Lu, Q. Zhang, L. Zheng, H. Wang, R.Y. Li, L.F. Zhang, W.B. Shen and X.D. Tu, Biol. Pharm. Bull., 27, 1515 (2004). 25. S.C. Chi and H.W. Jun, J. Pharm. Sci., 79, 974 (1990). 26. L.P. Ruan, B.Y. Yu, G.M. Fu and D. Zhu, J. Pharm. Biomed. Anal., 38, 457 (2005). 27. A. Figueiras, R.A. Carvalho, L. Ribeiro, J.J. Torres-Labandeira and F.J.B. Veiga, Eur. J. Pharm. Biopharm., 67, 531 (2007). (Received: 31 January 2008; Accepted: 3 November 2008) AJC-6984