Development of Polymeric Nanoparticles of Sunitinib malate in Eudragit S 100 for anti-colon cancer targeted drug delivery

Authors

DOI:

https://doi.org/10.59480/phahs.v1i2.30

Keywords:

Sunitinib, Eudragit S 100, nanoparticles, particle size, MTT assay, colon cancer

Abstract

  Sunitinib malate (SM) is a multi-targeted tyrosine kinase (TK) inhibitor. It is proposed as a potent anticancer drug intended for colon cancer. The current work aimed at developing Sunitinib malate -loaded nanoparticles for colon- targeting. SM loaded ES100-nanoparticles were developed by nano-precipitation method using poly vinyl alcohol (PVA) as stabilizer. Different formulation of ESNP (F1-F3) were prepared and evaluated for particle size, poly-dispersity index, and drug entrapment, using Fourier transform infrared spectroscopy (FTIR), x-ray diffraction (XRD), and scanning electron microscopy (SEM). The optimized formulation was further studied by in-vitro drug release and anticancer cell line study. The mean particle size, PDI, %EE of optimized SM loaded ESNPF2 was found to be 384.8±5.53 nm, 0.58, and 60.31% respectively. In vitro release of the optimized ESNPF2 showed 10.2 % drug release till 2 h, at 4h 29.6% and 12 h and 24 h 66.45%, 82.2% respectively at pH 7.4. SEM images and surface morphology confirmed that nanoparticles were spherical and had smooth surfaces. An MTT assay was conducted in the colon cancer cell lines (CACO2) to evaluate anti-cancerous activity of SM. SM loaded ESNPF2 showed a high potential against colon cancer cells. The overall results suggest that SM loaded ES100-nanoparticels could be a potential option for colon targeting.

References

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018; 68 (6):394–424. doi :10.3322/ caac.21492

Siegel RL, Miller KD, Goding Sauer A, et al. Colorectal cancer statistics, 2020. CA Cancer J Clin. 2020; 70(3):145–164. doi:10.3322/caac.21601

Brouwer NPM, Bos ACRK, Lemmens VEPP, et al. An overview of 25 years of incidence, treatment and outcome of colorectal cancer patients. Int J Cancer. 2018; 143(11):2758–2766. doi:10.1002/ijc.31785

Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 2009; 22(4):191–197. doi:10.1055/s-0029-1242458

Häfner MF, Debus J. Radiotherapy for colorectal cancer: current standards and future perspectives. Visc Med. 2016;32(3):172–177. doi:10.1159/000446486

Aiello P, Sharghi M, Mansourkhani SM, et al. Medicinal plants in the prevention and treatment of colon cancer. Oxid Med Cell Longev. 2019;2019:1–51. doi:10.1155/2019/2075614

Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global patterns and trends in colorectal cancer incidence and mortality. Gut. 2017;66 (4):683–691. doi:10.1136/gutjnl2015-310912

MendelD B, Laird AD, Xin X, Louie SG, Christensen JG, Li G, Schreck RE, Abrams TJ, Ngai TJ, Lee LB, Murray LJ, Carver J, Chan E, Moss KG, Haznedar JO, Sukbuntherng J, Blake RA, Sun L, Tang C, Miller T, Shirazian S, McMahon G, Cherrington JM. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin. Cancer Res. 2003; 9 :327–337.

Zheng X, Zhang Y, Zhang L, Xu W, Ma W, Sun R, Zeng H. Synergistic inhibition of sunitinib and ethaselen against human colorectal cancer cells proliferation. Biomed. Pharmacother. 2016; 83 :212–220.

Kwon H J, Kim Y, Sugihara Y, Baldetorp B, Welinder C, Watanabe K, Nishimura T, Malm J, Torok S, Dome B, Vegvari A, Gustavsson L, Fehniger TE, Marko-Varga G. Drug compound characterization by mass spectrometry imaging in cancer tissue. Arch. Pharm. Res. 2015; 38 :1718-1728.

Papaetis GS, Syrigos KN. Sunitinib: A multitargeted receptor tyrosine kinase inhibitor in the era of molecular cancer therapies. BioDrugs. 2009; 23-37

Ramazani F, Hiemstra C, Steendam R, Kazazi-Hyseni F, Van Nostrum CF, Storm G, Kiessling F, Lammers T, Hennink WE, Kok R.J. Sunitinib microspheres based on [PDLLA-PEG-PDLLA]-b-PLLA multi-block copolymers for ocular drug delivery. Eur. J. Pharm. Biopharm. 2015; 95:368–377.

Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D, Vinik A. et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N. Engl. J. Med. 2011; 364:501–513.

Nazari-Vanani R, Azarpira N, Heli H, Karimian K, Sattarahmady, N. A novel self-nanoemulsifying formulation for sunitinib: Evaluation of anticancer efficacy. Colloids Surfaces B Biointerfaces. 2017; 160:65–72.

Adams VR, Leggas M. Sunitinib malate for the treatment of metastatic renal cell carcinoma and gastrointestinal stromal tumors. Clin. Ther. 2007;29: 1338–1353.

Alshahrani SM, Alshetaili AS, Alalaiwe A, Alsulays BB, Anwer MK, Al-Shdefat R, Imam F, Shakeel F. Anticancer Efficacy of Self-Nanoemulsifying Drug Delivery System of Sunitinib Malate. AAPS PharmSciTech. 2018;19:123–133.

Chen S, Liang Q, Liu E, Yu Z, Sun L, Ye J, Shin M, Wang J, He H.

Curcumin/sunitinib co-loaded BSA-stabilized SPIOs for synergistic combination for breast cancer. J. Mater. Chem. B 2017; 5:4060–4072.

US EPA; Estimation Program Interface (EPI) Suite. Ver. 4.1. Jan, 2010. Available from, as of Apr 25, 2011.

The Merck Index ‐ An Encyclopedia of Chemicals, Drugs, and Biologicals. O'Neil, M.J. (ed.). Whitehouse Station, NJ: Merck and Co., Inc., 2006; p. 1544.

Gion PD, Kanefendt F, Lindauer A, Scheffler M, Doroshyenko O, Fuhr U, Wolf J, Jaehde U. Clinical pharmacokinetics of tyrosine kinase inhibitors. Clin. Pharmacokin. 2011; 50: 551-603.

Nimmons D, Limdi JK. Elderly patients and inflammatory bowel disease. World journal of gastrointestinal pharmacology and therapeutics. 2016; 7(1):51-53.

Abhishek SB, Parthiban S, Kumar GS, Mani TT. Microparticle as Suitable Drug Carriers for Colon Targeting-A Recent Review. American Journal of PharmTech Research. 2018; 8(1): 46-50

Amidon S, Brown JE, Dave VS. Colon-targeted oral drug delivery systems: design trends and approaches. AAPS Pharm Sci Tech. 2015; 16(4):731–741. doi:10.1208/s12249-015-0350-9

Pridgen EM, Alexis F, Farokhzad OC. Polymeric nanoparticle technologies for oral drug delivery. Clin Gastroenterol Hepatol. 2014;12(10):1605–1610. doi:10.1016/j.cgh.2014.06.018.

Hussan SD. A review on recent advances of enteric coating. IOSR J Pharm. 2012;2(6):05–11. doi:10.9790/3013-2610511

. Leopold CS. Coated dosage forms for colon-specific drug delivery. Pharm Sci Technol Today. 1999;2(5):197–204. doi:10.1016/S1461-5347(99)00151-0

Hao S, Wang B, Wang Y, Xu Y. Enteric-coated sustained-release nanoparticles by coaxial electrospray: preparation, characterization, and in vitro evaluation. J Nanoparticle Res. 2014;16 (2):2204. doi:10.1007/s11051-013-2204-2

Li C, Zhou K, Chen D, et al. Solid lipid nanoparticles with enteric coating for improving stability, palatability, and oral bioavailability of enrofloxacin. Int J Nanomedicine. 2019;14:1619–1631. doi:10.2147/IJN.S183479.

. Bhadra S, Prajapati A, Bhadra D. Development of pH sensitive polymeric nanoparticles of erythromycin stearate. J Pharm Bioallied Sci. 2016; 8 (2):135. doi:10.4103/0975-7406.17169

Tummala S, Kuppusamy G, Satish Kumar MN, Praveen TK, Wadhwani A. 5-Fluorouracil enteric-coated nanoparticles for improved apoptotic activity and therapeutic index in treating colorectal cancer. Drug Deliv. 2016;23(8):2902–2910. doi:10.3109/10717544.2015.1116026

Hosny KM. Alendronate sodium as enteric coated solid lipid nanoparticles; preparation, optimization, and in vivo evaluation to enhance its oral bioavailability. Santos HA, ed. PLoS One. 2016;11(5):e0154926. doi:10.1371/journal.pone.0154926

Valat MT. Mechanistic study of NVP-CGM097: A potent, selective and species specific inhibitor of p53-Mdm2. Drug Des Open Access. 2015;04:02. doi:10.4172/2169-0138.S1.008

Amini-Fazl MS, Mohammadi R, Kheiri K. 5-Fluorouracil loaded chitosan/polyacrylic acid/Fe 3 O 4 magnetic nanocomposite hydrogel as a potential anticancer drug delivery system. Int J Biol Macromol. 2019;132:506–513. doi:10.1016/j. ijbiomac.2019.04.005

Choi JS, Cao J, Naeem M, et al. Size-controlled biodegradable nanoparticles: preparation and size-dependent cellular uptake and tumor cell growth inhibition. Colloids Surfaces B Biointerfaces. 2014;122:545–551. doi:10.1016/j.colsurfb.2014.07.030

Choi YH, Han HK. Nanomedicines: current status and future perspectives in aspect of drug delivery and pharmacokinetics. J Pharm Investig. 2018;48(1):43–60. doi:10.1007/s40005-017-0370-4

. Banerjee A, Qi J, Gogoi R, Wong J, Mitragotri S. Role of nanoparticle size, shape and surface chemistry in oral drug delivery. J Control Release. 2016;238:176–185. doi:10.1016/j. jconrel.2016.07.051

Karn PR, Vanić Z, Pepić I, Škalko-Basnet N. Mucoadhesive liposomal delivery systems: the choice of coating material. Drug Dev Ind Pharm. 2011;37(4):482–488. doi:10.3109/ 03639045.2010.523425

Nikam V, Kotade K, Gaware V, et al. Eudragit a versatile polymer: a review. Pharmacologyonline. 2011;1:152–164.

Chen S, Guo F, Deng T, et al. Eudragit S100-coated chitosan nanoparticles co-loading tat for enhanced oral colon absorption of insulin. AAPS Pharm Sci Tech. 2017;18(4):1277–1287. doi:10.1208/s12249-016-0594-z

Ahmed M M, Anwer MK , Fatima F, Aldawsari M F, Alalaiwe A, Alali A S, Alharthi AI. and Kalam M A. Boosting the Anticancer Activity of Sunitinib Malate in Breast Cancer through Lipid Polymer Hybrid Nanoparticles Approach . Polymers. 2022; 14:2459. https://doi.org/10.3390/polym14122459

Alshetaili A S, Anwer M K, Alshahrani S M, Alalaiwe A, Alsulays BB, Ansari M J, Imam F , Alshehri S. Characteristics and anticancer properties of Sunitinib malate-loaded poly-lactic-co-glycolic acid nanoparticles against human colon cancer HT-29 cells lines. Tropical Journal of Pharmaceutical Research. 2018; 17 (7): 1263-1269

Sorasitthiyanukarn F N, Muangnoi C, Bhuket P R, Rojsitthisak P, Rojsitthisak P. Chitosan/alginate nanoparticles as a promising approach for oral delivery of curcumin diglutaric acid for cancer treatment. Mater. Sci. Eng. C Mater. Biol. Appl. 2018; (1)93:178-190. doi: 10.1016/j.msec.2018.07.069.

Anwer MK, Ahmed M M, Ezzeldin E, Fatima F, Alalaiwe A, Iqbal M. Preparation of sustained release apremilast-loaded PLGA nanoparticles: In vitro characterization and in vivo pharmacokinetic study in rats. Int. J. Nanomed. 2019; 14:1587–1595.

Jamil A, Aamir Mirza M, Anwer MK, Thakur PS, Alshahrani SM, Alshetaili AS, Telegaonkar S, Panda AK, Iqbal Z. Co-delivery of gemcitabine and simvastatin through PLGA polymeric nanoparticles for the treatment of pancreatic cancer: In-vitro characterization, cellular uptake, and pharmacokinetic studies. Drug Dev. Ind. Pharm. 2019; 45:745–753.

Anwer MK, Al-Shdefat R, Ezzeldin E, Alshahrani SM, Alshetaili AS, Iqbal M. Preparation, Evaluation and Bioavailability Studies of Eudragit Coated PLGA Nanoparticles for Sustained Release of Eluxadoline for the Treatment of Irritable Bowel Syndrome. Front. Pharm. 2017; 8: 844.

Y-Kırımlıoğlu G, Görgülü Ş & Berkman M S. Novel approaches to cancer therapy with ibuprofen-loaded Eudragit® RS 100 and/or octadecylamine-modified PLGA nanoparticles by assessment of their effects on apoptosis. Published online: 17 Jun 2020 https://doi.org/10.1080/03639045.2020.1776319

Barros S S, Silva J R, Azevedo R B, Lunardi C N & Gomes A J. Polyacrylate nanoparticles mediated cytotoxic process in breast cancer 4T1 cell line, MRS Online Proceedings Library. 2014; 1625: 619 Published: 01 April 2014

Y-Kırımlıoğlu G, Güleç K, Görgülü Ş, TubaKıyan H. Oseltamivir phosphate loaded pegylated-Eudragit nanoparticles for lung cancer therapy: Characterization, prolonged release, cytotoxicity profile, apoptosis pathways and in vivo anti-angiogenic effect by using CAM assay. Microvascular Research. 2022; 139:104251

Barichello JM, Morishita M, Takayama K, Nagai T. Encapsulation of hydrophilic and lipophilic drugs in PLGA nanoparticles by the nanoprecipitation method. Drug Dev Ind Pharm. 1999;25(4):471–476.

Fessi H, Puisieux F, Devissaguet JP, Ammoury N, Benita S. Nanocapsule formation by interfacial polymer deposition following solvent displacement. Int J Pharm. 1989;55:25–28.

Fessi H, Devissaguet JP, Puisieux F, Thies C. Process for the preparation of dispersible colloidal systems of a substance in the form of nanoparticles. 1992; US 593-522

Anwer MK, Al-Mansoor MA, Jamil S, Al-Shdefat R, Ansari MN, Shakeel F. Development and evaluation of PLGA polymer based nanoparticles of quercetin. Int J Biol Macromol 2016; 92: 213-219.

Josepha J J, Sangeethaa D, Gomathi T. Sunitinib loaded chitosan nanoparticles formulation and its evaluation. International Journal of Biological Macromolecules. 2016; 82:952–958

Andhya B, ChinnariHarika V, KasimallaB B, Syed R, Pammi K. RP-HPLC method development and validation for the analyisis of Sunitinib in pharmaceutical dosage forms. Int J Sci Innovations Discoveries 2011;1:441-50.

Blanchet B, Saboureau C, Benichou A-S, Billemont B, Taieb F, Ropert S, et al. Development and validation of an HPLC-UV-visible method for sunitinib quantification in human plasma. ClinChimActa 2009;404:134-9.

Konduri SKM, Satya BRAK, Chowdary NV. Polymorphic form of sunitinib base. United States Patent no. US 8,466,190 B2, 2013

Selic L. new crystal form of Sunitinib malate. European Patent Number EP2362873B1, 3 June 2015.

Published

2023-12-30

How to Cite

Jamil, S., Anwer , M. K., Sadeq, T., & Kohli , K. (2023). Development of Polymeric Nanoparticles of Sunitinib malate in Eudragit S 100 for anti-colon cancer targeted drug delivery. Pharmacy and Applied Health Sciences, 2(2), 20–29. https://doi.org/10.59480/phahs.v1i2.30

Issue

Section

Research Article