571
Views
35
CrossRef citations to date
0
Altmetric
Original Research

Ionically Cross-Linked Chitosan Nanoparticles for Sustained Delivery of Docetaxel: Fabrication, Post-Formulation and Acute Oral Toxicity Evaluation

, ORCID Icon, ORCID Icon, &
Pages 10035-10046 | Published online: 20 Dec 2019

References

  • WHO. Cancer; 2018 Available from: https://www.who.int/news-room/fact-sheets/detail/cancer. Accessed 912, 2018.
  • Siegel RL, Miller KD, Jemal A. Cancer statistics. CA Cancer J Clin. 2018;68(1):7–30. doi:10.3322/caac.2144229313949
  • Xu X, Ho W, Zhang X, Bertrand N, Farokhzad O. Cancer nanomedicine: from targeted delivery to combination therapy. Trends Mol Med. 2015;21(4):223–232. doi:10.1016/j.molmed.2015.01.00125656384
  • Bamrungsap S, Zhao Z, Chen T, et al. Nanotechnology in therapeutics: a focus on nanoparticles as a drug delivery system. Nanomedicine (Lond). 2012;7(8):1253–1271. doi:10.2217/nnm.12.8722931450
  • Acharya S, Sahoo SK. PLGA nanoparticles containing various anticancer agents and tumour delivery by EPR effect. Adv Drug Deliv Rev. 2011;63(3):170–183. doi:10.1016/j.addr.2010.10.00820965219
  • Moghimi SM, Hunter AC, Murray JC. Long-circulating and target-specific nanoparticles: theory to practice. Pharmacol Rev. 2001;53(2):283–318.11356986
  • Kumari A, Yadav SK, Yadav SC. Biodegradable polymeric nanoparticles based drug delivery systems. Colloids Surf B Biointerfaces. 2010;75(1):1–18. doi:10.1016/j.colsurfb.2009.09.00119782542
  • Qi L, Xu Z, Jiang X, Hu C, Zou X. Preparation and antibacterial activity of chitosan nanoparticles. Carbohydr Res. 2004;339(16):2693–2700. doi:10.1016/j.carres.2004.09.00715519328
  • Abd El Hady WE, Mohamed EA, Soliman OAE, El-Sabbagh HM. In vitro-in vivo evaluation of chitosan-PLGA nanoparticles for potentiated gastric retention and anti-ulcer activity of diosmin. Int J Nanomedicine. 2019;14:7191–7213. doi:10.2147/IJN.S21383631564873
  • Stenger Moura FC, Perioli L, Pagano C, et al. Chitosan composite microparticles: a promising gastroadhesive system for taxifolin. Carbohydr Polym. 2019;218:343–354. doi:10.1016/j.carbpol.2019.04.07531221339
  • Tsai L-C, Chen C-H, Lin C-W, Ho Y-C, Mi F-L. Development of multifunctional nanoparticles self-assembled from trimethyl chitosan and fucoidan for enhanced oral delivery of insulin. Int J Biol Macromol. 2019;126:141–150. doi:10.1016/j.ijbiomac.2018.12.18230586591
  • Malik A, Gupta M, Gupta V, Gogoi H, Bhatnagar R. Novel application of trimethyl chitosan as an adjuvant in vaccine delivery. Int J Nanomed. 2018;13:7959–7970. doi:10.2147/IJN
  • Anitha A, Deepagan VG, Divya Rani VV, Menon D, Nair SV, Jayakumar R. Preparation, characterization, in vitro drug release and biological studies of curcumin loaded dextran sulphate–chitosan nanoparticles. Carbohydr Polym. 2011;84(3):1158–1164. doi:10.1016/j.carbpol.2011.01.005
  • Cortes JE, Pazdur R. Docetaxel. J Clin Oncol. 1995;13(10):2643–2655. doi:10.1200/JCO.1995.13.10.26437595719
  • Jibodh RA, Lagas JS, Nuijen B, Beijnen JH, Schellens JHM. Taxanes: old drugs, new oral formulations. Eur J Pharmacol. 2013;717(1):40–46. doi:10.1016/j.ejphar.2013.02.05823660368
  • Jeetah R, Bhaw-Luximon A, Jhurry D. Polymeric nanomicelles for sustained delivery of anti-cancer drugs. Mutat Res. 2014;768:47–59. doi:10.1016/j.mrfmmm.2014.04.00924768976
  • Feng -S-S, Mei L, Anitha P, Gan CW, Zhou W. Poly(lactide)–vitamin E derivative/montmorillonite nanoparticle formulations for the oral delivery of docetaxel. Biomaterials. 2009;30(19):3297–3306. doi:10.1016/j.biomaterials.2009.02.04519299012
  • Thanki K, Gangwal RP, Sangamwar AT, Jain S. Oral delivery of anticancer drugs: challenges and opportunities. J Control Release. 2013;170(1):15–40. doi:10.1016/j.jconrel.2013.04.02023648832
  • Calvo P, Remunan‐Lopez C, Vila‐Jato JL, Alonso M. Novel hydrophilic chitosan‐polyethylene oxide nanoparticles as protein carriers. J Appl Polym Sci. 1997;63(1):125–132. doi:10.1002/(SICI)1097-4628(19970103)63:1<125::AID-APP13>3.0.CO;2-4
  • Rama K, Senapati P, Das M. Formulation and in vitro evaluation of ethyl cellulose microspheres containing zidovudine. J Microencapsul. 2005;22(8):863–876. doi:10.1080/0265204050027349816423758
  • Tahir N, Madni A, Balasubramanian V, et al. Development and optimization of methotrexate-loaded lipid-polymer hybrid nanoparticles for controlled drug delivery applications. Int J Pharm. 2017;533(1):156–168. doi:10.1016/j.ijpharm.2017.09.06128963013
  • Rehman M, Ihsan A, Madni A, et al. Solid lipid nanoparticles for thermoresponsive targeting: evidence from spectrophotometry, electrochemical, and cytotoxicity studies. Int J Nanomed. 2017;12:8325–8336. doi:10.2147/IJN
  • Twu Y-K, Chen Y-W, Shih C-M. Preparation of silver nanoparticles using chitosan suspensions. Powder Technol. 2008;185(3):251–257. doi:10.1016/j.powtec.2007.10.025
  • Bobal P, Sujan J, Otevrel J, Imramovsky A, Padelkova Z, Jampilek J. Microwave-assisted synthesis of new substituted anilides of quinaldic acid. Molecules. 2012;17(2):1292. doi:10.3390/molecules1702129222293847
  • Jain A, Thakur K, Kush P, Jain UK. Docetaxel loaded chitosan nanoparticles: formulation, characterization and cytotoxicity studies. Int J Biol Macromol. 2014;69:546–553. doi:10.1016/j.ijbiomac.2014.06.02924971551
  • Madni A, Rahim MA, Mahmood MA, et al. Enhancement of dissolution and skin permeability of pentazocine by proniosomes and niosomal gel. AAPS Pharm Sci Tech. 2018;19(4):1544–1553. doi:10.1208/s12249-018-0967-6
  • OECD G. 420. acute oral toxicity––fixed dose procedure. OECD Guidelines for the Testing of Chemicals; 2001.
  • Kleine-Brueggeney H, Zorzi GK, Fecker T, El Gueddari NE, Moerschbacher BM, Goycoolea FM. A rational approach towards the design of chitosan-based nanoparticles obtained by ionotropic gelation. Colloids Surf B Biointerfaces. 2015;135:99–108. doi:10.1016/j.colsurfb.2015.07.01626241921
  • Umair M, Javed I, Rehman M, et al. Nanotoxicity of inert materials: the case of gold, silver and iron. J Pharm Pharm Sci. 2016;19(2):161–180. doi:10.18433/J3102127518167
  • Alasvand N, Urbanska AM, Rahmati M, et al. Chapter 13 - therapeutic nanoparticles for targeted delivery of anticancer drugs In: Grumezescu AM, editor. Multifunctional systems for combined delivery, biosensing and diagnostics. Elsevier; 2017:245–259.
  • Wu J, Wang Y, Yang H, Liu X, Lu Z. Preparation and biological activity studies of resveratrol loaded ionically cross-linked chitosan-TPP nanoparticles. Carbohydr Polym. 2017;175:170–177. doi:10.1016/j.carbpol.2017.07.05828917853
  • Sreekumar S, Goycoolea FM, Moerschbacher BM, Rivera-rodriguez GR. Parameters influencing the size of chitosan-TPP nano- and microparticles. Sci Rep. 2018;8(1):4695. doi:10.1038/s41598-018-23064-429549295
  • Chang Y-C, Chen D-H. Preparation and adsorption properties of monodisperse chitosan-bound Fe3O4 magnetic nanoparticles for removal of Cu (II) ions. J Colloid Interface Sci. 2005;283(2):446–451. doi:10.1016/j.jcis.2004.09.01015721917
  • de Pinho Neves AL, Milioli CC, Müller L, Riella HG, Kuhnen NC, Stulzer HK. Factorial design as tool in chitosan nanoparticles development by ionic gelation technique. Colloids Surf a Physicochem Eng Asp. 2014;445:34–39. doi:10.1016/j.colsurfa.2013.12.058
  • Derakhshandeh K, Fathi S. Role of chitosan nanoparticles in the oral absorption of Gemcitabine. Int J Pharm. 2012;437(1):172–177. doi:10.1016/j.ijpharm.2012.08.00822909993
  • Hassani S, Laouini A, Fessi H, Charcosset C. Preparation of chitosan–TPP nanoparticles using microengineered membranes – effect of parameters and encapsulation of tacrine. Colloids Surf A Physicochem Eng Asp. 2015;482:34–43. doi:10.1016/j.colsurfa.2015.04.006
  • Nimesh S. Gene therapy: Potential applications of nanotechnology. 1st ed. Elsevier; 2013.
  • Yang W, Fu J, Wang T, He N. Chitosan/sodium tripolyphosphate nanoparticles: preparation, characterization and application as drug carrier. J Biomed Nanotechnol. 2009;5(5):591–595. doi:10.1166/jbn.2009.106720201437
  • Yuan D, Jacquier JC, O’Riordan ED. Entrapment of protein in chitosan-tripolyphosphate beads and its release in an in vitro digestive model. Food Chem. 2017;229:495–501. doi:10.1016/j.foodchem.2017.02.107.28372206
  • Jana S, Maji N, Nayak AK, Sen KK, Basu SK. Development of chitosan-based nanoparticles through inter-polymeric complexation for oral drug delivery. Carbohydr Polym. 2013;98(1):870–876. doi:10.1016/j.carbpol.2013.06.06423987423
  • Khanmohammadi M, Elmizadeh H, Ghasemi K. Investigation of size and morphology of chitosan nanoparticles used in drug delivery system employing chemometric technique. Iran J Pharm Res. 2015;14(3):665.26330855
  • Bilensoy E, Sarisozen C, Esendağlı G, et al. Intravesical cationic nanoparticles of chitosan and polycaprolactone for the delivery of Mitomycin C to bladder tumors. Int J Pharm. 2009;371(1–2):170–176. doi:10.1016/j.ijpharm.2008.12.01519135514
  • Zhu H, Chen H, Zeng X, et al. Co-delivery of chemotherapeutic drugs with vitamin E TPGS by porous PLGA nanoparticles for enhanced chemotherapy against multi-drug resistance. Biomaterials. 2014;35(7):2391–2400. doi:10.1016/j.biomaterials.2013.11.08624360574
  • Fang G, Tang B, Liu Z, et al. Novel hydrophobin-coated docetaxel nanoparticles for intravenous delivery: in vitro characteristics and in vivo performance. Eur J Pharm Sci. 2014;60:1–9. doi:10.1016/j.ejps.2014.04.01624815943
  • Hammadi NI, Abba Y, Hezmee MNM, et al. Formulation of a sustained release docetaxel loaded cockle shell-derived calcium carbonate nanoparticles against breast cancer. Pharm Res. 2017;34(6):1193–1203. doi:10.1007/s11095-017-2135-128382563
  • Nasrollahi F, Varshosaz J, Khodadadi AA, Lim S, Jahanian-Najafabadi A. Targeted delivery of docetaxel by use of transferrin/poly(allylamine hydrochloride)-functionalized graphene oxide nanocarrier. ACS Appl Mater Interfaces. 2016;8(21):13282–13293. doi:10.1021/acsami.6b0279027158834
  • Radwan SE-S, Sokar MS, Abdelmonsif DA, El-Kamel AH. Mucopenetrating nanoparticles for enhancement of oral bioavailability of furosemide: in vitro and in vivo evaluation/sub-acute toxicity study. Int J Pharm. 2017;526(1):366–379. doi:10.1016/j.ijpharm.2017.04.07228487189
  • Mukhopadhyay P, Chakraborty S, Bhattacharya S, Mishra R, Kundu PP. pH-sensitive chitosan/alginate core-shell nanoparticles for efficient and safe oral insulin delivery. Int J Biol Macromol. 2015;72:640–648. doi:10.1016/j.ijbiomac.2014.08.04025239194
  • Pati F, Adhikari B, Dhara S. Development of chitosan–tripolyphosphate fibers through pH dependent ionotropic gelation. Carbohydr Res. 2011;346(16):2582–2588. doi:10.1016/j.carres.2011.08.02821962591
  • Hejjaji EMA, Smith AM, Morris GA. Designing chitosan-tripolyphosphate microparticles with desired size for specific pharmaceutical or forensic applications. Int J Biol Macromol. 2017;95:564–573. doi:10.1016/j.ijbiomac.2016.11.09227894825
  • Bagre AP, Jain K, Jain NK. Alginate coated chitosan core shell nanoparticles for oral delivery of enoxaparin: in vitro and in vivo assessment. Int J Pharm. 2013;456(1):31–40. doi:10.1016/j.ijpharm.2013.08.03723994363
  • Bugnicourt L, Ladavière C. Interests of chitosan nanoparticles ionically cross-linked with tripolyphosphate for biomedical applications. Prog Polym Sci. 2016;60:1–17. doi:10.1016/j.progpolymsci.2016.06.002
  • Saremi S, Atyabi F, Akhlaghi SP, Ostad SN, Dinarvand R. Thiolated chitosan nanoparticles for enhancing oral absorption of docetaxel: preparation, in vitro and ex vivo evaluation. Int J Nanomed. 2011;6:119–128. doi:10.2147/IJN.S15500
  • Madni A, Sarfraz M, Rehman M, et al. Liposomal drug delivery: a versatile platform for challenging clinical applications. J Pharm Pharm Sci. 2014;17(3):401–426. doi:10.18433/J3CP5525224351
  • Sreejayan N, Marone PA, Lau FC, Yasmin T, Bagchi M, Bagchi D. Safety and toxicological evaluation of a novel chromium (III) dinicocysteinate complex. Toxicol Mech Methods. 2010;20(6):321–333. doi:10.3109/15376516.2010.48788020515439
  • Udeh NE, Anaga A, Asuzu IU. Acute and sub-chronic oral toxicity studies on methanol leaf extract of Gnetum africanum Welw in wistar rats. Am J Res Med Sci. 2018;3(1):7–14.
  • Raina P, Chandrasekaran C, Deepak M, Agarwal A, Ruchika K-G. Evaluation of subacute toxicity of methanolic/aqueous preparation of aerial parts of O. sanctum in Wistar rats: clinical, haematological, biochemical and histopathological studies. J Ethnopharmacol. 2015;175:509–517. doi:10.1016/j.jep.2015.10.01526456329
  • Mohammed MA, Syeda JTM, Wasan KM, Wasan EK. An overview of chitosan nanoparticles and its application in non-parenteral drug delivery. Pharmaceutics. 2017;9(4):53. doi:10.3390/pharmaceutics9040053