Studies on the cyclosporin A loaded stearic acid nanoparticles

Similar documents
FORMULATION AND CHARACTERIZATION OF TELMISATAN SOLID DISPERSIONS

7. SUMMARY, CONCLUSION AND RECOMMENDATIONS

Preparation and Characterization of Candesartan Cilexetil Solid Lipid Nanoparticulate Capsules

EFFECT OF COMPRESSED CO 2 ON THE PROPERTIES OF AOT IN ISOOCTANE REVERSE MICELLAR SOLUTION AND ITS APPLICATION TO RECOVER NANOPARTICLES

DEVELOPMENT OF SOLID LIPID NANOPARTICLES OF A WATER SOLUBLE DRUG

Formulation and Evaluation of Solid lipid nanoparticles: Isoniazid

3.1 Background. Preformulation Studies

Development and Characterization of Lipid Nanoparticles prepared by Miniemulsion Technique

COPYRIGHTED MATERIAL. Contents. xiv xv xvi. About the authors Preface Acknowledgments

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

Fluorescent Carbon Dots as Off-On Nanosensor for Ascorbic Acid

Pelagia Research Library

Formulation and Evaluation of Acyclovir Liposomes

Determination of Tetracyclines in Chicken by Solid-Phase Extraction and High-Performance Liquid Chromatography

Development of Nutrient Delivery Systems: Ingredients & Challenges

ENHANCEMENT OF SOLUBILITY OF BICALUTAMIDE DRUG USING SOLID DISPERSION TECHNIQUE

Supporting information for the manuscript

>>> Oral Formulation Optimization. Introduction. A Tiered Approach for Identifying Enabling Formulations

Tamoxifen Drug Loading Solid Lipid Nanoparticles Prepared by Hot High Pressure Homogenization Techniques

Lutein Esters from Tagetes Erecta

LABORATORY MANUAL PHM 2143 PHYSICAL PHARMACY II

THE INFLUENCE OF OILS AND SURFACTANTS ON THE FORMATION OF SELF-NANOEMULSIFYING DRUG DELIVERY SYSTEMS (SNEDDS) CONTAINING THERAPEUTIC PROTEIN

A study on the effects of different surfactants on Ethylcellulose microspheres

Bioequivalence Studies of Two Formulations of Famciclovir Tablets by HPLC Method

Supporting Information. Maximizing the Supported Bilayer Phenomenon: LCP Liposomes Comprised Exclusively

1. Gastric Emptying Time Anatomically, a swallowed drug rapidly reaches the stomach. Eventually, the stomach empties its content in the small

Insects as novel food ingredient

Optimisation and Stability Assessment of Solid Lipid Nanoparticles using Particle Size and Zeta Potential

Biopharmaceutics Dosage form factors influencing bioavailability Lec:5

FORMULATION CHOICE. How and why they are chosen. Dr Andy Fowles On behalf of ECPA Specification Expert Group

Thatipamula RP., Palem CR., Gannu R., Mudragada S., * Yamsani MR.

B. semisolid materials consisting of hydrophilic and hydrophobic portions

Measuring Lipid Composition LC-MS/MS

Determination of propranolol in dog plasma by HPLC method

Determination and pharmacokinetics of manidipine in human plasma by HPLC/ESIMS

1.2 Nano- or microencapsulation as a rich source of delivery of functional components 3

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

Study on Targeting and in vitro Anti-oxidation of Baicalin Solid Lipid Nanoparticles

Supporting Information

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

Determination of Gamma-Hydroxy-Butyrate (GHB) in Biological Samples

Formulation and Evaluation of Nanostructured Lipid Carrier (NLC) For Glimepiride

Electronic Supplementary Material

Electronic Supplementary Information

Naoya Takahashi, Keiya Hirota and Yoshitaka Saga* Supplementary material

Title Revision n date

Application Note. Authors. Abstract. Food

RESEARCH ARTICLE e-issn:

Effective use of Pharmacopeia guidelines to reduce cost of chromatographic analysis for Fluticasone propionate

Transparent Dispersions of Milk Fat-Based Solid Lipid Nanoparticles for Delivery of beta-carotene

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

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

STARCHES FOR COSMETIC INDUSTRIES CORN PO4 PH B AND RICE NS

DEVELOPMENT OF IN VITRO-IN VIVO CORRELATION FOR ENCAPSULATED METOPROLOL TARTRATE

Determination of β2-agonists in Pork Using Agilent SampliQ SCX Solid-Phase Extraction Cartridges and Liquid Chromatography-Tandem Mass Spectrometry

Mass-Based Purification of Natural Product Impurities Using an Agilent 1260 Infinity II Preparative LC/MSD System

PREPARATION AND CHARACTERIZATION OF LORNOXICAM LOADED SOLID LIPID NANOPARTICLES MADE FROM DIFFERENT LIPIDS

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

Liposomes in polymer matrix. Stability of liposomes in PEG 400 and PEG 8000 solutions.

4. Monitoring of dissolution induced changes in film coat

Extraction and Characterization of Galactomannan from Guar Seeds

An Orthogonal Array Optimization of Lipid-like Nanoparticles for. mrna Delivery in Vivo

A high-performance liquid chromatography (HPLC) method for determination of chlorogenic acid and emodin in Yinhuang Jiangzhi Tea

Sulfate Radical-Mediated Degradation of Sulfadiazine by CuFeO 2 Rhombohedral Crystal-Catalyzed Peroxymonosulfate: Synergistic Effects and Mechanisms

INTERNATIONAL JOURNAL OF PHARMACY & LIFE SCIENCES

The preparation of silybin phospholipid complex and the study on its pharmacokinetics in rats

Fatty Acid Methylation Kits

Design and Optimization of Rivaroxaban Lipid Solid Dispersion for Dissolution Enhancement using Statistical Experimental Design. ), and Labrasol (X 3

Osnove farmakokinetike. Aleš Mrhar. Prirejeno po. A First Course in Pharmacokinetics and Biopharmaceutics by David Bourne,

Transdermal Delivery of Newer Atypical Antipsychotics ABSTRACT

Journal of Pharmaceutical and Scientific Innovation

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

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

Heparin Sodium ヘパリンナトリウム

J Pharm Sci Bioscientific Res (4): ISSN NO

Preparation, Characterization and In vivo Evaluation of Rosuvastatin Calcium Loaded Solid Lipid Nanoparticles

Supporting Information for. A Lipophilic Pt(IV) Oxaliplatin Derivative Enhances Antitumor activity

2- Minimum toxic concentration (MTC): The drug concentration needed to just produce a toxic effect.

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

In vitro In vivo correlation of sustained release capsules of Metoprolol

EUDRAGIT L 100 and EUDRAGIT S 100

4. Determination of fat content (AOAC, 2000) Reagents

Encapsulation techniques

Product Guide for LudgerSep TM ur2 UHPLC Column for DMB Sialic Acid Analysis

Jagua (Genipin-Glycine) Blue (Tentative)

Product Guide for LudgerSep TM R1 HPLC Column for DMB labelled Sialic Acid Analysis

Isolation of five carotenoid compounds from tangerine tomatoes

ANALYSIS OF -HYDROXYBUTYRATE (GHB) AND -BUTYROLACTONE (GBL) IN LIQUIDS PERFORMED AT NATIONAL LABORATORY OF FORENSIC SCIENCE (SKL), SWEDEN

Journal of Chemical and Pharmaceutical Research, 2018, 10(1): Research Article

THIN LAYER CHROMATOGRAPHY

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

IJRPC 2011, 1(4) Rohan et al. ISSN: INTERNATIONAL JOURNAL OF RESEARCH IN PHARMACY AND CHEMISTRY

Analysis of HMF by HPLC

EFFECT OF SURFACE MODIFICATION OF SOLID LIPID NANOPARTICLES IN BRAIN TARGETING

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

EFFICIENT PRODUCTION OF SOY-BEAN LECITHIN PLURONIC L64 ENCAPSULATED QUERCETIN PARTICLES IN NANOMETRIC SCALE USING SFEE AND PGSS DRYING PROCESSES

International Journal of Food Nutrition and Safety, 2012, 1(2): International Journal of Food Nutrition and Safety

[ APPLICATION NOTE ] Profiling Mono and Disaccharides in Milk and Infant Formula Using the ACQUITY Arc System and ACQUITY QDa Detector

PRODUCTION OF VALUE ADDED PRODUCTS FROM SOME CEREAL MILLING BY-PRODUCTS SAYED SAAD ABOZAIED SMUDA THESIS DOCTOR OF PHILOSOPHY

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

Transcription:

International Journal of Pharmaceutics 200 (2000) 153 159 www.elsevier.com/locate/ijpharm Studies on the cyclosporin A loaded stearic acid Qiang Zhang a, Guoqing Yie a, Yie Li a, Qingsong Yang a, T. Nagai b, * a Department of Pharmacy, School of Pharmaceutical Sciences, Beijing Medical Uni ersity, Beijing 100083, People s Republic of China b Department of Pharmaceutics, Hoshi Uni ersity, Ebara 2-4-41, Shinagawa-ku, Tokyo 142, Japan Received 27 September 1999; received in revised form 8 November 1999; accepted 15 December 1999 Abstract Stearic acid were prepared in this study by melt-homogenization to investigate the possibility of them as a new kind of drug carrier system. Some physicochemical properties of stearic acid were studied and morphology examined by transmission electron microscope. Cyclosporin A as a model drug was then encapsulated into stearic acid. Following the establishment of high performance liquid chromatography assay for cyclosporin A analysis in stearic acid or blood samples, the encapsulation ratio of cyclosporin A to stearic acid was estimated and pharmacokinetics as well as bioavailability of cyclosporin A stearic acid after oral administration to Wistar rats were studied, using the Sandimmun Neoral (an available microemulsion system of cyclosporin A) as a reference. The mean diameter of cyclosporin A stearic acid was 316.1 nm, while the encapsulation ratio of cyclosporin A to stearic acid reached to 88.36%. It was demonstrated by IR spectra and differential scanning calorimetry that there was no chemical reaction occurred between the cyclosporin A and stearic acid. The relative bioavailability of cyclosporin A stearic acid over reference was nearly 80%, and the time to reach maximum concentration (T max ) of cyclosporin A after oral administration of cyclosporin A stearic acid was delayed significantly than the reference, suggesting an obvious sustained release effect. The stearic acid might be a very potential drug carrier. 2000 Published by Elsevier Science B.V. All rights reserved. Keywords: Drug carrier; Stearic acid; Nanoparticles; Cyclosporin A; Relative bioavailability 1. Introduction Particulate drug delivery system, such as and microspheres has been studied * Corresponding author. Tel.: +81-3-59400385; fax: +81-3-59400386. E-mail address: nagai@hoshi.ac.jp (T. Nagai) extensively in the past 10 years. But the toxicity of the carrier materials was still remained to be problems since the synthesized polymers such as alkylcyanoacrylate, poly(lactic acid), methylmethacrylate, and so on were often used. The possible accumulation and their toxic metabolic product were still worth to be further studied. 0378-5173/00/$ - see front matter 2000 Published by Elsevier Science B.V. All rights reserved. PII: S0378-5173(00)00361-6

154 Q. Zhang et al. / International Journal of Pharmaceutics 200 (2000) 153 159 In order to choice a better carrier material, an endogenous longchain saturated fatty acid (stearic acid) was studied in this investigation. The stearic acid is a main composition of fat, so it was expected to have better biocompatibility and lower toxicity than the synthesized polymers. Because it is solid at room temperature it was supposed to be more stable than liposome or emulsion. Stearic acid is available for pharmaceutical use and it is easy to prepare the with it. Several approaches on preparation (Schwarz and Wehnert, 1997; Freitas and Muller, 1998), stability (Heiati, et al., 1998; Freitas and Muller, 1999) and other aspects (Bargoni et al., 1998; Morel et al., 1998) have been reported recently about solid lipid. The purpose of our present investigation was to evaluate the possibility of stearic acid as a new kind of drug carrier, through the studies of preparation procedure, physicochemical properties, quality as well as the pharmacokinetics and bioavailability of cyclosporin A (model drug) stearic acid after oral administration to rats. 2. Materials and methods 2.1. Materials Cyclosprione A was obtained from Sichuan Industrial Institute of Antibiotics (Chengdu, China). Cyclosprione D was a gift from Northern-China Pharmaceutical Plant (Shijiazhuang, China). Stearic acid was the product of Shanghai People s Chemical Plant (Shanghai, China). Lecithin and Poloxomer were purchased from Sigma (St Louis, MO). Sandimmun Neoral (microemulsion system of cyclosporin A) was the product of Sandoz (Sweden). Methanol and acetonitrile were purchased from Beijing People s Chemical Plant (Beijing, China). All other chemicals and solvents were of analytical grade. 2.2. Preparation ofstearic acid Stearic acid were prepared by melt-homogenization (Siekmann and Westesen, 1994). Briefly, the aqueous solution containing surfactants was heated to 75 C, then under mechanical stirring the solution was added to stearic acid melted at the same temperature. The stirring was kept to form stearic acid until room temperature. 2.3. Physicochemical properties ofstearic acid The morphology of stearic acid was examined by transmission electron microscope. Zeta potential was measured by an U- tube electrophoresis apparatus, surface tension by maximum bubble method, turbidity by recording the absorption at 600 nm, relative density by an areometer, viscosity by an Ubbelohde viscometer, ph value by a ph-25 acidimeter and refractive index by an Abbe refractometer. 2.4. Preparation of cyclosporin A stearic acid Cyclosporin A stearic acid were obtained by the same approach mentioned above, except that cyclosporin A and stearic acid were melted together. 2.5. Encapsulation ratio of cyclosporin A to stearic acid Cyclosporin A stearic acid were separated from the liquid medium by centrifuging (60 000 rpm, 2 h). The sediment obtained was dissolved in methanol, then analyzed at 55 C in a high performance liquid chromatography system with an Alltech C 18 column (5 m, 250 mm 4.6 mm). The samples were eluted with methanol:water (90:10) at a flow rate of 1.0 ml/ min and detected at 214 nm. Encapsulation ratio (ER) of cyclosporin A to stearic acid was calculated from: ER=(W m /W a ) 100%, where W m represents the amount of cyclosporin A found while W a means the amount of cyclosporin A added.

Q. Zhang et al. / International Journal of Pharmaceutics 200 (2000) 153 159 155 2.6. Particle size of cyclosporin A stearic acid The colloidal solution of cyclosporin A stearic acid was diluted with distilled water, then analyzed in a particle size analyzer. 2.7. IR spectra and differential scanning calorimetry analysis The IR spectra of cyclosporin A, freeze-dried stearic acid and freeze-dried cyclosporin A stearic acid were prepared in a IR spectrometer. The differential scanning calorimetry thermograms of stearic acid, the mixture of cyclosporin A and stearic acid, and the freeze-dried cyclosporin A stearic acid were made by a differential scanning calorimeter. 2.8. Analysis of cyclosporin A in blood samples Cyclosporin A in blood samples was analyzed at 75 C by a high performance liquid chromatography system (Liang and Wu, 1992) with an Hypersil 120 C 18 packed column (5 m, 150 mm 4.6 mm). The samples were eluted with acetonitrile:methanol:water (60:10:30) at a flow rate of 1.5 ml/min and detected at 210 nm. Cyclosporin D was taken as the internal material. 2.9. Pharmacokinetics and bioa ailability of cyclosporin A stearic acid in rats Cyclosporin A stearic acid (the test) were compared with the reference, Sandimmun Neoral. Ten 250 25 g Wistar rats (Animal Center of Beijing Medical University), fasted overnight, were matched in pairs according to the body weight. After appropriate dilution, the single dose of cyclosporin A stearic acid or reference was given intragastrically to the rat in each pair, respectively. Venous blood were collected from the postorbital vein sinus before and over a 60-h period, then the plasma samples were separated and kept at 4 C until analysis. The elimination rate constant (K el ) was calculated with log-linear regression, using the last three to five observations of each curve, and the elimination half-life (t 1/2 ) was derived from 0.693/ K el. The area under the plasma concentration time curve (AUC 0 t ) was calculated with the trapezoidal rule, and extrapolation of the area to infinity (AUC 0 inf ) was estimated by adding the last measured plasma concentration divided by the elimination rate constant (K el ). A computer program 3P87 (Administration of Health, Beijing, China) was used for the fitting of pharmacokinetic models, and also for the calculation of the maximum plasma concentration (C max ) as well as the time to attain C max (t max ). The relative bioavailability (Fr%) was calculated from (AUC 0 inf for test /AUC 0 inf for reference ) 100%. 3. Results Fig. 1. Micrograph of stearic acid by transmission electron microscope ( 14 000). The colloidal solution of stearic acid prepared was white and milk-like. Most of the looked round and regular under transmission electron microscope as shown in Fig. 1. Their physicochemical properties of stearic acid were listed in Table 1. The stearic acid were negatively charged, but the zeta potential was rather low. The surface tension and ph value of stearic acid decreased, while the viscosity and turbidity increased, as the concentration of stearic acid increased. Little change was observed in relative density and refractive index.

156 Q. Zhang et al. / International Journal of Pharmaceutics 200 (2000) 153 159 Table 1 Physicochemical properties of stearic acid (n=5) Physicochemical properties Concentration of SA-NP a 1.2% 2.0% 3.0% Zeta potential (mv) 3.45 0.19 2.60 0.12 4.02 0.26 Surface tension (10 2,N/m) 5.32 0.09 4.75 0.05 3.80 0.04 Turbidity 0.037 0.001 0.091 0.002 0.2090 0.001 Relative density (g/cm 3 ) 1.0015 0.0001 1.0013 0.0001 1.0016 0.0002 Viscosity (10 3,Ns/m 2 ) 1.404 0.002 1.473 0.003 1.933 0.003 ph value 4.28 0.06 3.93 0.02 3.82 0.02 Refractive index 1.3388 0.0001 1.3389 0.0001 1.3393 0.0001 a SA-NP, stearic acid. The encapsulation ratio of cyclosporin A to stearic acid reached to 88.36% as shown in Table 2, analyzed by HPLC method. The linear response of this assay ranged from 0.5 to 200 g/ml. The analytical recoveries were 97.82, 100.03 and 100.71%, respectively, under three different concentration (5.0, 25.0 and 100.0 g/ ml), while the relative standard deviation within day were 0.54, 0.56 and 1.62%, respectively. The mean diameter of cyclosporin A stearic acid was 316.1 nm, and 84.4% of the cyclosporin A stearic acid distributed between 176 and 297 nm. It was demonstrated by IR spectra in Fig. 2 that there was no chemical reaction occurred between the cyclosporin A and stearic acid, because the characteristic peaks of cyclosporin A and stearic acid did not show any shifts after encapsulation of cyclosporin A to stearic acid. Studies on differential scanning calorimetry came to the same conclusion. The baseline separation between cyclosporin A and cyclosporin D was achieved with the high performance liquid chromatography system established for analysis of cyclosporin A in blood samples. The standard curves were linear over the range of 50 1000 ng/ml, and the retention time of cyclosporin A and D were 6.96 and 9.16 min, respectively. The detective limitation was 20 ng/ ml. The analytical recoveries were 111.7, 101.1 and 100.8%, respectively under three different concentration (50, 300 and 1000 ng/ml), while the relative standard deviation within day were 5.3, 3.6 and 2.7%, respectively. Cyclosporin A plasma concentrations (mean S.D.) for both single dose oral administration of cyclosporin A stearic acid and the reference were shown in Fig. 3, and the derived pharmacokinetic parameters in Table 3. A rapid increase followed by a rapid decrease in cyclosporin A plasma concentrations resulted after oral administration of microemulsion system Table 2 Encapsulation ratio of cyclosporin A to stearic acid (n=3) No. CyA a added ( g/ml) CyA found ( g/ml) Encapsulation ratio % Mean % 1 27.85 24.67 88.69 88.36 2.19 2 26.38 22.74 86.28 3 27.45 24.64 90.11 a CyA, cyclosporin A.

Q. Zhang et al. / International Journal of Pharmaceutics 200 (2000) 153 159 157 Table 3 Pharmacokinetic parameters of cyclosporin A after single dose oral administration of cyclosporin A stearic acid or Sandimmun Neoral to Wistar rats Pharmacokinetic CyA SA-NP a Sandimmun Neoral parameter (1.2325 mg/100 g) (1.0 mg/100 g) C max (ng/ml) 1033.1 353.2 1736.4 358.0 t max (h) 4.5 2.2 1.75 0.43 AUC 0 60 22.77 3.36 23.27 3.43 ( g h/ml) AUC 0 inf ( g h/ml) 25.04 3.16 25.47 3.26 K el (1/h) 0.0453 0.0122 0.0464 0.0177 T 1/2 (h) 16.42 4.16 16.90 5.29 Fr (%) 79.77 a CyA SA-NP, cyclosporin A stearic acid. Fig. 2. The IR spectra of cyclosporin A (upper), stearic acid (middle) and cyclosporin A stearic acid (down). of cyclosporin A, with C max occurring at about 1.7 h. Relatively slow increase and also slow decrease in cyclosporin A plasma concentrations were observed after administration of cyclosporin A stearic acid, with the C max of cyclosporin A occurring at 4.0 h which is delayed significantly (P 0.05) than the reference, suggesting an obvious sustained release effect of it. The pharmacokinetic parameters such as K el and t 1/2 shown no statistically significant difference (P 0.05) between the two treatments. The relative bioavailability of cyclosporin A stearic acid over reference was Fig. 3. Mean plasma concentration of cyclosporin A vs. time curves after single oral administration of cyclosporin A stearic acid ( ) or Sandimmun Neoral ( ) to rats.

158 Q. Zhang et al. / International Journal of Pharmaceutics 200 (2000) 153 159 nearly 80%, indicating that stearic acid could improve the absorption of some insoluble drug, such as cyclosporin A, in the gastrointestinal tract. The cyclosporin A plasma concentration time curve after single oral administration of cyclosporin A stearic acid was well described by biexponential equation according to a one-compartment open model, while the cyclosporin A plasma concentration time data was well fitted by a two-compartment model with one-order absorption kinetics. 4. Discussion The preparation techniques investigated in this work included solvent-evaporation and melt-homogenization, and the sonication dispersion and mechanical dispersion had been used in melthomogenization. The stearic acid prepared by solvent-evaporation could be very small and unique but always aggregated to form precipitation, while the obtained by sonication dispersion at high temperature were irregular and rather big in diameter. Mechanical dispersion from high temperature to room temperature was proved to be the best method in the preparation of stearic acid. Small, unique and stable tearic acid could be prepared with this technique. Effects of stirring time, stirring speed and temperature on the particle size and morphology were studied and the best formulation was selected. It was demonstrated that stirring time and speed show no obvious affects on the particle size and its homogeneity, and the present of cyclosporin A had no influence on the formation of. It was observed in the established HPLC system that the temperature show great impact on the column efficiency, and the column efficiency increased with the rise of temperature. A temperature of 55 C was selected for the analysis temperature for analysis of cyclosporin A in stearic acid in consideration of column damage at high temperature. But higher temperature was employed for analysis of cyclosporin A in blood samples because more impurities existed and baseline separation of cyclosporin A and D was needed. The Sandimmun Neoral, an available microemulsion system of cyclosporin A was used as a reference in the studies of pharmacokinetics and bioavailability of cyclosporin A stearic acid after oral administration. As a new product of Sandoz, the microemulsion system, made of surfactants and oil phase, can form emulsion droplets only about 30 nm whenever it contact with water, so it is specially advantageous for the absorption of cyclosporin A, which is a typical poor soluble drug. Even compared with such formulation, the relative bioavailability of cyclosporin A stearic acid was nearly 80%, indicated that stearic acid could improve the absorption of cyclosporin A. The major reason of lower bioavailability of cyclosporin A stearic acid might be the size of the (316.1 nm) which is tenfold of the microemulsion droplets ( 30 nm), and other possibility may due to the different surfactants used. It was noticed in Fig. 3 that the profiles of the cyclosporin A plasma concentrations after single dose oral administration of cyclosporin A stearic acid and Sandimmun Neoral were quite different. It could be attributed to the different release rate of different delivery systems, because the insoluble cyclosporin A is rather ready to release from the liquid microemulsion droplets than from the solid. The effect of food and particle size on the absorption of cyclosporin A in GI tract was remained to be further studied. References Bargoni, A., Cavalli, R., Caputo, O., et al., 1998. Solid lipid in lymph and plasma after duodenal administration to rats. Pharm. Res. 15, 745 750. Freitas, X., Muller, R.H., 1998. Spray-drying of solid lipid (SLN TM). Eur. J. Pharm. Biopharm. 46, 145 151. Freitas, X., Muller, R.H., 1999. Correlation between longterm stability of solid lipid (SLN) and crystallinity of the lipid phase. Eur. J. Pharm. Biopharm. 47, 125 132.

Q. Zhang et al. / International Journal of Pharmaceutics 200 (2000) 153 159 159 Heiati, H., Tawashi, R., Phillips, N.C., 1998. Drug retention and stability of solid lipid nanoparticies containing azidothymidine palmitate after autoclaving, storage and lyophilization. J. Microencapsul. 15, 173 184. Liang, Y., Wu, L.W., 1992. Measurement of cyclosporine in whole blood by high perforance liquid chromatography. Chin. J. Med. Lab. Tech. 15, 206 211. Morel, L.S., Terreno, E., Ugazio, E., et al., 1998. NMR relaxometric investigations of solid lipid (SLN) containing gadolinium(iii) complexes. Eur. J. Pharm. Biopharm. 45, 157 163. Schwarz, C., Wehnert, W., 1997. Freeze-drying of drug-free and drug-loaded solid lipid (SLN). Int. J. Pharm. 157, 171 179. Siekmann, B., Westesen, K., 1994. Thermoanalysis of the recrystallization process of melt-homogenized glyceride. Colloids Surf. B Biointerfaces 3, 159 175..