Research Article | Volume: 8, Issue: 10, October, 2018

DOE, formulation, and optimization of Repaglinide nanostructured lipid carriers

Shady A. Swidan Zeinab N. Mansour Zeinab A. Mourad Nahla A. Elhesaisy Nada A. Mohamed Mohamed S. Bekheet Mohamed A. Badawy Mai M. Elsemeiri Aya E. Elrefaey Amera M. Hassaneen   

Open Access   

Published:  Oct 31, 2018

DOI: 10.7324/JAPS.2018.81002
Abstract

Nanostructured lipid carriers (NLC) are a recent approach for the delivery of poorly soluble drugs with low oral bioavailability. The oral antidiabetic Repaglinide (RPG) was loaded into NLC using emulsification–ultrasonification technique. A design of experiment was constructed to study the formulation variables. The influence of the liquid lipid to the solid lipid ratio and the concentration of the surfactant on mean particle size, zeta potential, and drug entrapment efficiency was demonstrated. The mean particle size ranged from 182 ± 7.9 nm to 452 ± 66.1 nm. All particles were negatively charged and the zeta potential values ranged from −7.9 ± 0.9 mV to −44.4 ± 6.2 mV. The highest entrapment efficiency was obtained with the minimum solid lipid to liquid lipid ratio and lowest surfactant concentration. All RPG–NLC formulae showed biphasic time-dependent in vitro release and the studied factors were optimized and the optimum formula was evaluated for in vitro release and crystallinity. The in vitro release of the optimized formula fitted to the Higuchi diffusion model. In conclusion, this study showed the potential of NLC as a carrier for controlled release of RPG.


Keyword:     Nanostructured lipid carriers emulsification– ultrasonification Repaglinide optimization DSC.


Citation:

Swidan SA, Mansour ZN, Mourad ZA, Elhesaisy NA, Mohamed NA, Bekheet MS, et al. DOE, formulation, and optimization of Repaglinide nanostructured lipid carriers. J App Pharm Sci, 2018; 8(10): 008-016.

Copyright: © The Author(s). This is an open-access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

HTML Full Text

Reference

Abdelbary G, Haider G. In vitro characterization and growth inhibition effect of nanostructured lipid carriers for controlled delivery of methotrexate. Pharm Dev Technol, 2011; 18(5):1159–68. https://doi.org/10.3109/10837450.2011.614251

Akhtar J, Fareed S, Aqil M. Stability-indicating assay of repaglinide in bulk and optimized nanoemulsion by validated high performance thin layer chromatography technique. J Pharm Bioallied Sci, 2013; 5(3):184. https://doi.org/10.4103/0975-7406.116800

Akhtar J, Siddiqui HH, Fareed S, Badruddeen MK, Aqil M. Nanoemulsion: for improved oral delivery of repaglinide. Drug Deliv, 2016; 23(6):2026–34. https://doi.org/10.3109/10717544.2015.1077290

Chalikwar SS, Belgamwar VS, Talele VR, Surana SJ, Patil MU. Formulation and evaluation of nimodipine-loaded solid lipid nanoparticles delivered via lymphatic transport system. Colloids Surf B Biointerfaces, 2012; 97:109–16. https://doi.org/10.1016/j.colsurfb.2012.04.027

Das S, Chaudhury A. Recent advances in lipid nanoparticle formulations with solid matrix for oral drug delivery. AAPS PharmSciTech, 2011; 12(1):62–76. https://doi.org/10.1208/s12249-010-9563-0

Das S, Ng WK, Kanaujia P, Kim S, Tan RBH. Formulation design, preparation and physicochemical characterizations of solid lipid nanoparticles containing a hydrophobic drug: effects of process variables. Colloids Surf B Biointerfaces, 2011; 88(1):483–89. https://doi.org/10.1016/j.colsurfb.2011.07.036

Dubey A, Prabhu P, Kamath JV. Nano structured lipid carriers: a novel topical drug delivery system. Int J PharmTech Res, 2011; 4(2):705–14.

Ebrahimi HA, Javadzadeh Y, Hamidi M, Jalali MB. Repaglinide-loaded solid lipid nanoparticles: effect of using different surfactants/ stabilizers on physicochemical properties of nanoparticles. Daru, 2015; 23(1):46. https://doi.org/10.1186/s40199-015-0128-3

Gadadare R, Mandpe L, Pokharkar V. Ultra rapidly dissolving Repaglinide nanosized crystals prepared via bottom-up and top-down approach: influence of food on pharmacokinetics behavior. AAPS PharmSciTech, 2015; 16(4):787–99. https://doi.org/10.1208/s12249-014-0267-8

He W, Wu M, Huang S, Yin L. Matrix tablets for sustained release of Repaglinide: preparation, pharmacokinetics and hypoglycemic activity in Beagle dogs. Int J Pharm 2015; 478(1):297–307. https://doi.org/10.1016/j.ijpharm.2014.11.059

Honary S, Zahir F. Effect of zeta potential on the properties of nano-drug delivery systems—a review (Part 1). Trop J Pharm Res 2013; 12(2):255–64.

Hu S, Wang S, Fanelli B, Bell PA, Dunning BE, Geisse S, et al. Pancreatic beta-cell K (ATP) channel activity and membrane-binding studies with nateglinide: a comparison with sulfonylureas and Repaglinide. J Pharmacol Exp Ther, 2000; 293(2):444–52.

Kamble MS, Borwandkar VG, Bodade SS, Aute PP, Chaudhari PD, Bhosale AV. Optimization of self-nanoemulsifying drug delivery system (SNEDDS) of Repaglinide using D-optimal mixture experimental design. J Biomed Pharm Res, 2013; 2(3):100–8, 109.

Kassem, AA, Abd El-Alim SH, Basha M, Salama A. Phospholipid complex enriched micelles: a novel drug delivery approach for promoting the antidiabetic effect of Repaglinide. Eur J Pharm Sci, 2017; 99:75–84. https://doi.org/10.1016/j.ejps.2016.12.005

Kramer CK, Zinman B, Gross JL, Canani LH, Rodrigues TC, Azevedo MJ, et al. Coronary artery calcium score prediction of all cause mortality and cardiovascular events in people with type 2 diabetes: systematic review and meta-analysis. BMJ, 2013; 346(1):1654–4. https://doi.org/10.1136/bmj.f1654

Kumar GSS, Talsania MP, Goli D, Karki R. Formulation and optimization of nanostructured lipid matrices of Repaglinide using factorial design. World J Pharm Pharm Sci, 2013; 2(6):5521–37.

Luo YF, Chen DW, Ren LX, Zhao XL, Qin J. Solid lipid nanoparticles for enhancing vinpocetine's oral bioavailability. J Control Release, 2006; 114(1):53–9. https://doi.org/10.1016/j.jconrel.2006.05.010

Maiti K, Mukherjee K, Gantait A, Saha BP, Mukherjee PK. Curcumin-phospholipid complex: preparation, therapeutic evaluation and pharmacokinetic study in rats. Int J Pharm, 2007; 330(1–2):155–63. https://doi.org/10.1016/j.ijpharm.2006.09.025

Mazumder S, Dewangan AK, Pavurala N. Enhanced dissolution of poorly soluble antiviral drugs from nanoparticles of cellulose acetate based solid dispersion matrices. Asian J Phar Sci, 2017; 12(6):532–41. https://doi.org/10.1016/j.ajps.2017.07.002

Purvis T, Mattucci ME, Crisp MT, Johnston KP, Williams RO. Rapidly dissolving Repaglinide powders produced by the ultra-rapid freezing process. AAPS PharmSciTech, 2017; 8(3):E58.

Sanad RA, Abdelmalak NS, Elbayoomy TS, Badawi AA. Formulation of a novel oxybenzone-loaded nanostructured lipid carriers (NLCs). AAPS PharmSciTech, 2010; 11(4):1684–94. https://doi.org/10.1208/s12249-010-9553-2

Sangsen Y, Laochai P, Chotsathidchai P, Wiwattanapatapee R. Effect of solid lipid and liquid oil ratios on properties of nanostructured lipid carriers for oral curcumin delivery. Adv Mat Res, 2014; 1060:62–5. https://doi.org/10.4028/www.scientific.net/AMR.1060.62

Siepmann J, Peppas NA. Higuchi equation: derivation, applications, use and misuse. Int J Pharm, 2011; 418(1):6–12. https://doi.org/10.1016/j.ijpharm.2011.03.051

Swidan SA, Ghonaim HM, Samy AM, Ghorab MM. Efficacy and in vitro cytotoxicity of nanostructured lipid carriers for paclitaxel delivery. J Appl Pharm Sci, 2016; 6(9):018–26. https://doi.org/10.7324/JAPS.2016.60903

Tan SW, Billa N, Roberts CR, Burley JC. Surfactant effects on the physical characteristics of amphotericin B-containing nanostructured lipid carriers. Colloids Surf A Physicochem Eng Aspects, 2010; 372(1–3):73–9. https://doi.org/10.1016/j.colsurfa.2010.09.030

Teeranachaideekul V, Boonme P, Souto EB, Müller RH, Junyaprasert VB. Influence of oil content on physicochemical properties and skin distribution of nile red-loaded NLC. J Control Release, 2008; 128(2):134–41. https://doi.org/10.1016/j.jconrel.2008.02.011

Teeranachaideekul V, Souto EB, Junyaprasert VB, Müller RH. Cetyl palmitate-based NLC for topical delivery of coenzyme Q10— development, physicochemical characterization and in vitro release studies. Eur J Pharm Biopharm, 2007; 67(1):141–8. https://doi.org/10.1016/j.ejpb.2007.01.015

Venkateswarlu V, Manjunath K. Preparation, characterization and in vitro release kinetics of clozapine solid lipid nanoparticles. J Control Release, 2004; 95(3):627–38. https://doi.org/10.1016/j.jconrel.2004.01.005

Vera Candioti L, De Zan MM, Cámara MS, Goicoechea HC. Experimental design and multiple response optimization. Using the desirability function in analytical methods development. Talanta, 2014; 124:123–38. https://doi.org/10.1016/j.talanta.2014.01.034

Weber S, Zimmer A, Pardeike J. Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) for pulmonary application: a review of the state of the art. Eur J Pharm Biopharm, 2014; 86(1):7–22. https://doi.org/10.1016/j.ejpb.2013.08.013

Yang Y, Corona A, Schubert B, Reeder R, Henson MA. The effect of oil type on the aggregation stability of nanostructured lipid carriers. J Colloid Interface Sci, 2014; 418:261–72. https://doi.org/10.1016/j.jcis.2013.12.024

Yu H, Huang Q. Bioavailability and delivery of nutraceuticals and functional foods using nanotechnology. Bio-Nanotechnol Revol Food Biomed Health Sci, 2013; 593–604. https://doi.org/10.1002/9781118451915.ch35

Zeng N, Hu Q, Liu Z, Gao X, Hu R, Song Q, et al. Preparation and characterization of paclitaxel-loaded DSPE-PEG-liquid crystalline nanoparticles (LCNPs) for improved bioavailability. Int J Pharm, 2012; 424(1–2):58–66. https://doi.org/10.1016/j.ijpharm.2011.12.058

Zhu H, Zhang X, Li M-Z, Xie J, Yang X-L. Prevalence of type 2 diabetes and pre-diabetes among overweight or obese children in Tianjin, China. Diab Med J Br Diab Assoc, 2013; 30(12):1457–65. https://doi.org/10.1111/dme.12269

Article Metrics
999 Views 160 Downloads 1159 Total

Year

Month

Related Search

By author names