期刊文献+

Glucose-conjugated chitosan nanoparticles for targeted drug delivery and their specific interaction with tumor cells 被引量:3

Glucose-conjugated chitosan nanoparticles for targeted drug delivery and their specific interaction with tumor cells
原文传递
导出
摘要 A novel targeted drug delivery system, glucose-conjugated chitosan nanoparticles (GCNPs), was developed for specific recognition and interaction with glucose transporters (Gluts) over-expressed by tumor cells. GC was synthesized by using succinic acid as a linker between glucosamine and chitosan (CS), and successful synthesis was confirmed by NMR and elemental analysis. GCNPs were prepared by ionic crosslinking method, and characterized in terms of morphology, size, and zeta potential. The optimally prepared nanoparticles showed spherical shapes with an average particle size of (187.9 ± 3.8) nm and a zeta potential of (-15.43 ± 0.31) mV. The GCNPs showed negligible cytotoxicity to mouse embryo fibroblast and 4T1 cells. Doxorubicin (DOX) could be efficiently entrapped into GCNPs, with a loading capacity and encapsulation efficiency of 20.11% and 64.81%, respectively. DOX-Ioaded nanoparticles exhibited sustained-release behavior in phosphate buffered saline (pH 7.4). In vitro cellular uptake studies showed that the GCNPs had better endocytosis ability than CSNPs, and the antitumor activity of DOX/GCNPs was 4-5 times effectiveness in 4T1 cell killing than that of DOX/CSNPs. All the results demonstrate that nanoparticles decorated with glucose have specific interactions with cancer cells via the recognition between glucose and Gluts. Therefore, Gluts-targeted GCNPs may be promising delivery agents in cancer therapies. A novel targeted drug delivery system, glucose-conjugated chitosan nanoparticles (GCNPs), was developed for specific recognition and interaction with glucose transporters (Gluts) over-expressed by tumor cells. GC was synthesized by using succinic acid as a linker between glucosamine and chitosan (CS), and successful synthesis was confirmed by NMR and elemental analysis. GCNPs were prepared by ionic crosslinking method, and characterized in terms of morphology, size, and zeta potential. The optimally prepared nanoparticles showed spherical shapes with an average particle size of (187.9 ± 3.8) nm and a zeta potential of (-15.43 ± 0.31) mV. The GCNPs showed negligible cytotoxicity to mouse embryo fibroblast and 4T1 cells. Doxorubicin (DOX) could be efficiently entrapped into GCNPs, with a loading capacity and encapsulation efficiency of 20.11% and 64.81%, respectively. DOX-Ioaded nanoparticles exhibited sustained-release behavior in phosphate buffered saline (pH 7.4). In vitro cellular uptake studies showed that the GCNPs had better endocytosis ability than CSNPs, and the antitumor activity of DOX/GCNPs was 4-5 times effectiveness in 4T1 cell killing than that of DOX/CSNPs. All the results demonstrate that nanoparticles decorated with glucose have specific interactions with cancer cells via the recognition between glucose and Gluts. Therefore, Gluts-targeted GCNPs may be promising delivery agents in cancer therapies.
出处 《Frontiers of Materials Science》 SCIE CSCD 2014年第4期363-372,共10页 材料学前沿(英文版)
基金 This research was supported by the National Natural Science Foundation of China (Grant Nos. 31000423 and 31301420) and the China Postdoctoral Science Foundation (Grant No. 2014M551965).
关键词 drug delivery target nanoparticle glucose transporter (Glut) chitosan(CS) drug delivery target nanoparticle glucose transporter (Glut) chitosan(CS)
  • 相关文献

参考文献27

  • 1Chen C, Yu C H, Cheng Y C, et al. Biodegradable nanoparticles of amphiphilic triblock copolymers based on poly(3-hydroxybuty- rate) and poly(ethylene glycol) as drug carriers. Biomaterials, 2006, 27(27): 4804-4814.
  • 2Haley B, Frenkel E. Nanoparticles for drug delivery in cancer treatment. Urologic Oncology, 2008, 26(1): 57-64.
  • 3Malam Y, Loizidou M, Seifalian A M. Liposomes and nanoparticles: nanosized vehicles for drug delivery in cancer. Trends in Pharmacological Sciences, 2009, 30(11): 592-599.
  • 4Byrne J D, Betancourt T, Brannon-Peppas L. Active targeting schemes for nanoparticle systems in cancer therapeutics. Advanced Drug Delivery Reviews, 2008, 60(15): 1615-1626.
  • 5Sudimack J, Lee R J. Targeted drug delivery via the folate receptor. Advanced Drug Delivery Reviews, 2000, 41(2): 147- 162.
  • 6Hruz P W, Mueckler M M. Structural analysis of the GLUT1 facilitative glucose transporter. Molecular Membrane Biology, 2001, 18(3): 183-193.
  • 7Olson A L, Pessin J E. Structure, function, and regulation of the mammalian facilitative glucose transporter gene family. Annual Review of Nutrition, 1996, 16(1): 235-256.
  • 8Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. The Journal of General Physiology, 1927, 8(6): 519-530.
  • 9Cullinane C, Solomon B, Hicks R J. Imaging of molecular target modulation in oncology: challenges of early clinical trials. Clinical and Translational Imaging, 2014, 2(1): 5-12.
  • 10Liu D Z, Sinchaikul S, Reddy P V G, et al. Synthesis of 2'- paclitaxel methyl 2-glucopyranosyl succinate for specific targeted delivery to cancer cells. Bioorganic & Medicinal Chemistry Letters, 2007, 17(3): 617-620.

同被引文献17

引证文献3

二级引证文献7

相关作者

内容加载中请稍等...

相关机构

内容加载中请稍等...

相关主题

内容加载中请稍等...

浏览历史

内容加载中请稍等...
;
使用帮助 返回顶部