2,603
Views
17
CrossRef citations to date
0
Altmetric
Research Articles

Formulation, characterization, in vitro and in vivo evaluations of self-nanoemulsifying drug delivery system of luteolin

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 1386-1401 | Received 27 Sep 2019, Accepted 13 Aug 2020, Published online: 26 Sep 2020

References

  • Manzoor MF, Ahmad N, Manzoor A, et al. Food based phytochemical luteolin their derivatives, sources and medicinal benefits. Int J Agric Life Sci. 2017;3(2):195–207.
  • Gutiérrez-Grijalva EP, Picos-Salas MA, Leyva-López N, et al. Flavonoids and phenolic acids from oregano: occurrence, biological activity and health benefits. Plants (Basel). 2017;7(1):1–23.
  • Horváthová K, Novotný L, Tóthová D, et al. Determination of free radical scavenging activity of quercetin, rutin, luteolin and apigenin in H2O2-treated human ML cells K562. Neoplasma. 2004;51:395–399.
  • Qiusheng Z, Yuntao Z, Rongliang Z, et al. Effects of verbascoside and luteolin on oxidative damage in brain of heroin treated mice. Pharmazie. 2005;60:539–543.
  • Wölfle U, Esser PR, Simon-Haarhaus B, et al. UVB-induced DNA damage, generation of reactive oxygen species, and inflammation are effectively attenuated by the flavonoid luteolin in vitro and in vivo. Free Radic Biol Med. 2011;50:1081–1093.
  • Wölfle U, Haarhaus B, Schempp CM. The photoprotective and antioxidative properties of luteolin are synergistically augmented by tocopherol and ubiquinone. Planta Med. 2013;79:963–965.
  • Kimata M, Inagaki N, Nagai H. Effects of luteolin and other flavonoids on IgE-mediated allergic reactions. Planta Med. 2000;66:25–29.
  • Ueda H, Yamazaki C, Yamazaki M. Luteolin as an anti-inflammatory and anti-allergic constituent of Perillafrutescens. Biol Pharm Bull. 2002;25:1197–1202.
  • Hirano T, Higa S, Arimitsu J, et al. Flavonoids such as luteolin, fisetin and apigenin are inhibitors of interleukin-4 and interleukin-13 production by activated human basophils. Int Arch Allergy Immunol. 2004;134:135–140.
  • Baolin L, Weiwei W, Ning T. Topical application of luteolin inhibits scratching behavior associated with allergic cutaneous reaction in mice. Planta Med. 2005;71:424–428.
  • Kritas SK, Saggini A, Varvara G, et al. Luteolin inhibits mast cell-mediated allergic inflammation. J BiolReg Homeost Agents. 2013;27:955–959.
  • Fei J, Liang B, Jiang C, et al. Luteolin inhibits IL-1β-induced inflammation in rat chondrocytes and attenuates osteoarthritis progression in a rat model. Biomed Pharmacother. 2019;109:1586–1592.
  • Fan X, Du K, Li N, et al. Evaluation of anti-nociceptive and anti-inflammatory effect of luteolin in mice. J Environ Pathol Toxicol Oncol. 2018;37:351–364.
  • Rakariyatham K, Wu X, Tang Z, et al. Synergism between luteolin and sulforaphane in anti-inflammation. Food Funct. 2018;9:5115–5123.
  • Aziz N, Kim MY, Cho JY. Anti-inflammatory effects of luteolin: a review of in vitro, in vivo, and in silico studies. J Ethnopharmacol. 2018;225:342–358.
  • Xiong J, Wang K, Yuan C, et al. Luteolin protects mice from severe acute pancreatitis by exerting HO-1-mediated anti-inflammatory and antioxidant effects. Int J Mol Med. 2017;39:113–125.
  • Xia N, Chen G, Liu M, et al. Anti-inflammatory effects of luteolin on experimental autoimmune thyroiditis in mice. Exp Ther Med. 2016;12:4049–4054.
  • Kanai K, Nagata S, Hatta T, et al. Therapeutic anti-inflammatory effects of luteolin on endotoxin-induced uveitis in Lewis rats. J Vet Med Sci. 2016;78:1381–1384.
  • Yang PW, Lu ZY, Pan Q, et al. MicroRNA-6809-5p mediates luteolin-induced anticancer effects against hepatoma by targeting flotillin 1. Phytomedicine 2019;57:18–29.
  • Pu Y, Zhang T, Wang J, et al. Luteolin exerts an anticancer effect on gastric cancer cells through multiple signaling pathways and regulating miRNAs. J Cancer. 2018;9:3669–3675.
  • Palko-Labuz A, Sroda-Pomianek K, Uryga A, et al. Anticancer activity of baicalein and luteolin studied in colorectal adenocarcinoma LoVo cells and in drug-resistant LoVo/Dx cells. Biomed Pharmacother. 2017;88:232–241.
  • Lee YJ, Lim T, Han MS, et al. Anticancer effect of luteolin is mediated by downregulation of TAM receptor tyrosine kinases, but not interleukin-8, in non-small cell lung cancer cells. Oncol Rep. 2017;37:1219–1226.
  • Ma L, Peng H, Li K, et al. Luteolin exerts an anticancer effect on NCI-H460 human non-small cell lung cancer cells through the induction of Sirt1-mediated apoptosis. Mol Med Rep. 2015;12:4196–4202.
  • Yan J, Wang Q, Zheng X, et al. Luteolin enhances TNF-related apoptosis-inducing ligand’s anticancer activity in a lung cancer xenograft mouse model. Biochem Biophys Res Commun. 2012;417:842–846.
  • Zang Y, Igarashi K, Li Y. Anti-diabetic effects of luteolin and luteolin-7-O-glucoside on KK-A(y) mice. Biosci Biotechnol Biochem. 2016;80:1580–1586.
  • Choi JS, Islam MN, Ali MY, et al. The effects of C-glycosylation of luteolin on its antioxidant, anti-Alzheimer’s disease, anti-diabetic, and anti-inflammatory activities. Arch Pharm Res. 2014;37:1354–1363.
  • Kim JS, Kwon CS, Son KH. Inhibition of alpha-glucosidase and amylase by luteolin, a flavonoid. Biosci Biotechnol Biochem. 2000;64:2458–2461.
  • Shakeel F, Haq N, Alshehri S, et al. Solubility, thermodynamic properties and solute-solvent molecular interactions of luteolin in various pure solvents. J Mol Liq. 2018;255:43–50.
  • Product information Luteolin LKT LABS. Available from: https://www.lktlabs.com/product/luteolin/.
  • Lipinski CALF. Poor aqueous solubility – an industry wide problem in drug discovery. Am Pharm Rev. 2002;5:82–88.
  • Chen Z, Tu M, Sun S, et al. The exposure of luteolin is much lower than that of apigenin in oral administration of FlosChrysanthemi extract to rats. Drug Metab Pharmacokinet. 2012;27:162–168.
  • Lin LC, Pai YF, Tsai TH. Isolation of luteolin and luteolin-7-O-glucoside from Dendranthemamorifolium Ramat Tzvel and their pharmacokinetics in rats. J Agric Food Chem. 2015;63:7700–7706.
  • Liu B, Li W, Zhao J, et al. Physicochemical characterization of the supramolecular structure of luteolin/cyclodextrin inclusion complex. Food Chem. 2013;141:900–906.
  • Jullian C, Cifuentes C, Alfaro M, et al. Spectroscopic characterization of the inclusion complexes of luteolin with native and derivatized beta-cyclodextrin. Bioorg Med Chem. 2010;18:5025–5031.
  • Khan J, Saraf S, Saraf S. Preparation and evaluation of luteolin-phospholipid complex as an effective drug delivery tool against GalN/LPS induced liver damage. Pharm Dev Technol. 2016;21:475–486.
  • Khan J, Alexander A, Saraf S, et al. Luteolin-phospholipid complex: preparation, characterization and biological evaluation. J Pharm Pharmacol. 2014;66:1451–1462.
  • Xu K, Liu B, Ma Y, et al. Physicochemical properties and antioxidant activities of luteolin-phospholipid complex. Molecules. 2009;14:3486–3493.
  • Wu G, Li J, Yue J, et al. Liposome encapsulated luteolin showed enhanced antitumor efficacy to colorectal carcinoma. Mol Med Rep. 2018;17:2456–2464.
  • Zhao G, Zang SY, Jiang ZH, et al. Postischemic administration of liposome-encapsulated luteolin prevents against ischemia-reperfusion injury in a rat middle cerebral artery occlusion model. J Nutr Biochem. 2011;22:929–936.
  • Li J, Cheng X, Chen Y, et al. Vitamin E TPGS modified liposomes enhance cellular uptake and targeted delivery of luteolin: an in vivo/in vitro evaluation. Int J Pharm. 2016;512:262–272.
  • Huang M, Su E, Zheng F, et al. Encapsulation of flavonoids in liposomal delivery systems: the case of quercetin, kaempferol and luteolin. Food Funct. 2017;8:3198–3208.
  • Zheng S, Cheng Y, Teng Y, et al. Application of luteolin nanomicelles anti-glioma effect with improvement in vitro and in vivo. Oncotarget. 2017;8:61146–61162.
  • Qiu JF, Gao X, Wang BL, et al. Preparation and characterization of monomethoxypoly(ethylene glycol)-poly(ε-caprolactone) micelles for the solubilization and in vivo delivery of luteolin. Int J Nanomed. 2013;8:3061–3069.
  • Yan H, Wei P, Song J, et al. Enhanced anticancer activity in vitro and in vivo of luteolin incorporated into long-circulating micelles based on DSPE-PEG2000 and TPGS. J Pharm Pharmacol. 2016;68:1290–1298.
  • Qing W, Wang Y, Li H, et al. Preparation and characterization of copolymer micelles for the solubilization and in vitro release of luteolin and luteoloside. AAPS PharmSciTech. 2017;18:2095–2101.
  • Majumdar D, Jung KH, Zhang H, et al. Luteolin nanoparticle in chemoprevention: in vitro and in vivo anticancer activity. Cancer Prev Res. 2014;7:65–73.
  • Weerapol Y, Limmatvapirat S, Nunthanid J, et al. Self-nanoemulsifying drug delivery system of nifedipine: impact of hydrophilic-lipophilic balance and molecula structure of mixed surfactants. AAPS PharmSciTech. 2014;15:456–464.
  • Yoo JH, Shanmugam S, Thapa P, et al. Novel self-nanoemulsifying drug delivery system for enhanced solubility and dissolution of lutein. Arch Pharm Res. 2010;33:417–426.
  • El-Badry M, Haq N, Fetih G, et al. Solubility and dissolution enhancement of tadalafil using self-nanoemulsifying drug delivery system. J Oleo Sci. 2014;63:567–576.
  • Shakeel F, Haq N, Alanazi FK, et al. Thermodynamic modeling for solubility prediction of indomethacin in self-nanoemulsifying drug delivery system (SNEDDS) and its individual components. Drug Dev Ind Pharm. 2014;40:1240–1245.
  • Dash RN, Mohammed H, Humaira T, et al. Design, optimization and evaluation of glipizide solid self-nanoemulsifying drug delivery for enhanced solubility and dissolution. Saudi Pharm J. 2015;23:528–540.
  • Gamal W, Fahmy RH, Mohamed MI. Development of novel amisulpride-loaded liquid self-nanoemulsifying drug delivery systems via dual tackling of its solubility and intestinal permeability. Drug Dev Ind Pharm. 2017;43:1530–1538.
  • Kim DH, Maharjan P, Kim JY, et al. Enhanced solubility, in-vitro dissolution and lipase inhibition of a self-nanoemulsifying drug delivery system containing orlistat. J Nanosci Nanotechnol. 2019;19:634–639.
  • Karasulu HY, Gundogdu E, Turk UO, et al. Enhancing solubility and bioavailability of rosuvastatin into self-nanoemulsifying drug delivery system. Curr Drug Deliv. 2018;15:1072–1082.
  • Mohsin K, Alamri R, Ahmad A, et al. Development of self-nanoemulsifying drug delivery systems for the enhancement of solubility and oral bioavailability of fenofibrate, a poorly water-soluble drug. Int J Nanomed. 2016;11:2829–2838.
  • Larsen AT, Ohlsson AG, Polentarutti B, et al. Oral bioavailability of cinnarizine in dogs: relation to SNEDDS droplet size, drug solubility and in vitro precipitation. Eur J Pharm Sci. 2013;48:339–350.
  • Kim RM, Jang DJ, Kim YC, et al. Flurbiprofen-loaded solid SNEDDS preconcentrate for the enhanced solubility, in-vitro dissolution and bioavailability in rats. Pharmaceutics. 2018;10:E247.
  • Kim DH, Kim JY, Kim RM, et al. Flurbiprofen-loaded solid SNEDDS preconcentrate for the enhanced solubility, in-vitro dissolution and bioavailability in rats. Orlistat-loaded solid SNEDDS for the enhanced solubility, dissolution, and in vivo performance. Int J Nanomed. 2018;13:7095–7106.
  • Abouhussein DMN, Bahaa El Din Mahmoud D, Mohammad FE. Design of a liquid nano-sized drug delivery system with enhanced solubility of rivaroxaban for venous thromboembolism management in paediatric patients and emergency cases. J Liposome Res. 2019;1:1–14.
  • Ansari MJ, Ahmed MM, Anwer MK, et al. Formulation and characterization of fluconazole loaded olive oil nano-emulsions. Indo Am J Pharm Sci. 2017;4:852–860.
  • Louis D. Formulation and evaluation of nanocrystals of a lipid lowering agent. Iran J Pharm Res. 2016;15(1):71–82.
  • Kazi M, Al-Swairi M, Ahmad A, et al. Evaluation of self-nanoemulsifying drug delivery system (SNEDDS) for poorly water-soluble talinolol: preparation, in vitro and in vivo assessment. Front Pharmacol. 2019;10:459.
  • Saritha D, Bose P, Nagaraju R. Formulation and evaluation of self-emulsifying drug delivery system (SEDDS) of ibuprofen. Int J Pharm Sci Res. 2014;5:3511–3519.
  • Nasr A, Gardouh A, Ghorab M. Novel solid self-nanoemulsifying drug delivery system (S-SNEDDS) for oral delivery of olmesartanmedoxomil: design, formulation, pharmacokinetic and bioavailability evaluation. Pharmaceutics. 2016;8:E20.
  • Ansari MJ, Alshahrani SM. Nano-encapsulation and characterization of baricitinib using poly-lactic-glycolic acid co-polymer. Saudi Pharm J. 2019;27:491–501.
  • Khan AW, Kotta S, Ansari SH, et al. Self-nanoemulsifying drug delivery system (SNEDDS) of the poorly water-soluble grapefruit flavonoid naringenin: design, characterization, in vitro and in vivo evaluation. Drug Deliv. 2015;22:552–561.
  • AlShahrani S, Ansari MJ. Solubility evaluations of osimertinib mesylate in physiological buffers. Indo Am J Pharm Sci. 2018;5:2610–2615.
  • Kumar A, Ahuja A, Ali J, et al. Curcumin-loaded lipid nanocarrier for improving bioavailability, stability and cytotoxicity against malignant glioma cells. Drug Deliv. 2016;23:214–229.
  • Ruan LP, Chen S, Yu BY, et al. Prediction of human absorption of natural compounds by the non-everted rat intestinal sac model. Eur J Med Chem. 2006;41:605–610.
  • Reddy SA, Sen SA, Chakraborty RA, et al. Effect of gokhru plant extract on intestinal absorption of aspirin using everted sac technique. Int J Pharm Boil Arch. 2011;2:549–553.
  • Lassoued MA, Khemiss F, Sfar S. Comparative study of two in vitro methods for assessing drug absorption: Sartorius SM 16750 apparatus versus everted gut sac. J Pharm Pharm Sci. 2011;14(1):117–127.
  • Marinova G, Batchvarov V. Evaluation of the methods for determination of the free radical scavenging activity by DPPH. Bulg J Agric Sci. 2011;17(1):11–24.
  • Quest Graph™ IC50 Calculator (v.1). AAT Bioquest, Inc. Available from: https://www.aatbio.com/tools/ic50-calculator-v1.
  • Astata 2016, by Navendu Vasavada. Available from: https://astatsa.com/OneWay_Anova_with_TukeyHSD/_get_data/.
  • Ansari MJ, Kohli K, Ali J, et al. Enhanced anti-inflammatory activity of curcumin, a naturally occurring pigment in turmeric via cyclodextrin complexation. Orient Pharm Exp Med. 2007;7:121–127.
  • Jelveghari M, Nokhodchi A. Development and chemical stability studies of alcohol-free phenobarbital solution for use in pediatrics: a technical note. AAPS Pharm Sci Tech. 2008;9:939–943.
  • Censi R, Di Martino P. Polymorph impact on the bioavailability and stability of poorly soluble drugs. Molecules. 2015;20:18759–18776.
  • Kalepu S, Nekkanti V. Insoluble drug delivery strategies: review of recent advances and business prospects. Acta Pharm Sin B. 2015;5:442–453.
  • Kale SN, Deore SL. Emulsion micro emulsion and nano emulsion: a review. Syst Rev Pharm. 2017;8:39–47.
  • Emerenciano DP, Baracho BB, Medeiros ML, et al. Physicochemical characterizations and antioxidant property of copaiba oil loaded into SNEDDS systems. J Braz Chem Soc. 2019;30(2):234–246.
  • Beg S, Swain S, Singh HP, et al. Development, optimization, and characterization of solid self-nanoemulsifying drug delivery systems of valsartan using porous carriers. AAPS Pharm Sci Tech. 2012;13(4):1416–1427.
  • Kassem AA, Mohsen AM, Ahmed RS, et al. Self-nanoemulsifying drug delivery system (SNEDDS) with enhanced solubilization of nystatin for treatment of oral candidiasis: design, optimization, in vitro and in vivo evaluation. J Mol Liq. 2016;218:219–232.
  • Subramanian G, Meyyanathan S, Byran G. Stability-indicating reverse-phase high-performance liquid chromatography method for the simultaneous quantification of apigenin and luteolin from Achillea millefolium Linn. Asian J Pharm Clin Res. 2019;12(4):169–172.
  • Wang M, Jin X, Ren X, et al. Comparative in vitro dissolution of two commercially available Er-Zhi-Wan herbal medicinal products. Indian J Pharm Sci. 2015;77(4):391–398.
  • Ghai D, Sinha VR. Nanoemulsions as self-emulsified drug delivery carriers for enhanced permeability of the poorly water-soluble selective β1-adrenoreceptor blocker Talinolol. Nanomedicine. 2012;8(5):618–626.
  • Malik P, Tapan KM. Structure-function elucidation of antioxidative and prooxidative activities of the polyphenolic compound curcumin. Chin J Biol. 2014;396708.
  • Djeridane A, Yousfi M, Nadjemi B, et al. Screening of some Algerian medicinal plants for the phenolic compounds and their antioxidant activity. Eur Food Res Techn. 2007;224(6):801–809.
  • Amic D, Davidovic-Amic D, Beslo D. Trinajstic N: structure-radical scavenging activity relationship of flavonoids. Croat Chem Acta. 2003;76:55–61.
  • de Queiroz FR, Avaca LA. Electrochemical determination of the antioxidant capacity: the ceric reducing/antioxidant capacity (CRAC) assay. Electroanalysis. 2008;20(12):1323–1329.
  • de Queiroz FR, Avaca LA. Electrochemical determination of the antioxidant capacity of Brazilian woods as alternative materials for the aging of cachaça. Braz J Food Technol. 2008;4:27–33.
  • Olugbami JO, Gbadegesin MA, Odunola OA. In vitro free radical scavenging and antioxidant properties of ethanol extract of Terminalia glaucescens. Pharmacog Res. 2015;7(1):49–56.
  • Furusawa M, Tsuchiya H, Nagayama M, et al. Anti-platelet and membrane-rigidifying flavonoids in brownish scale of onion. J Health Sci. 2003;49:475–480.
  • De Martino L, Mencherini T, Mancini E, et al. In vitro phytotoxicity and antioxidant activity of selected flavonoids. Int J Mol Sci. 2012;13(5):5406–5419.
  • Furusawa M, Tanaka T, Ito T, et al. Antioxidant activity of hydroflavonoids. J Health Sci. 2005;51:376–378.
  • Baloch J, Sohail MF, Sarwar HS, et al. Self-nanoemulsifying drug delivery system (SNEDDS) for improved oral bioavailability of chlorpromazine: in vitro and in vivo evaluation. Medicina (B Aires). 2019;55(5):210–218.
  • Morales G, Paredes A, Olivares A, et al. Acute oral toxicity and anti-inflammatory activity of hydroalcoholic extract from Lampayamedicinalis Phil in rats. Biol Res. 2014;47(1):6–11.
  • Gambhire MN, Wankhede SS, Juvekar AR. Antiinflammatory activity of aqueous extract of Barleriacristata leaves. J Young Pharm. 2009;1(3):220–227.
  • Kanai K, Nagata S, Hatta T, et al. Therapeutic anti-inflammatory effects of luteolin on endotoxin-induced uveitis in Lewis rats. J Vet Sci. 2016: 16–19.
  • Fan X, Du K, Li N, et al. Evaluation of anti-nociceptive and anti-inflammatory effect of luteolin in mice. J Environ Pathol Toxicol Oncol. 2018;37(4):351–364.
  • Lodhi S, Vadnere GP, Patil KD, et al. Protective effects of luteolin on injury induced inflammation through reduction of tissue uric acid and pro-inflammatory cytokines in rats. J Tradit Complement Med. 2020;10(1):60–69.
  • Lamy S, Moldovan PL, Saad AB, et al. Biphasic effects of luteolin on interleukin-1β-induced cyclooxygenase-2 expression in glioblastoma cells. BBA-Mol Cell Res. 2015;1853(1):126–135.
  • Taha EI, Al-Suwayeh SA, El-Badry M. Bioavailability study of indomethacin self-nanoemulsifying oral formulation in rats. Aust J Basic Appl Sci. 2009;3(3):2944–2948.