DOI QR코드

DOI QR Code

Docetaxel-loaded PLGA nanoparticles to increase pharmacological sensitivity in MDA-MB-231 and MCF-7 breast cancer cells

  • 투고 : 2021.04.12
  • 심사 : 2021.05.17
  • 발행 : 2021.09.01

초록

This study aimed to develop docetaxel (DTX) loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (DTX-NPs) and to evaluate the different pharmacological sensitivity of NPs to MCF-7 and MDA-MB-231 breast cancer cells. NPs containing DTX or coumarin-6 were prepared by the nanoprecipitation method using PLGA as a polymer and d-α-tocopherol polyethylene glycol 1000 succinate (TPGS) as a surfactant. The physicochemical properties of NPs were characterized. In vitro anticancer effect and cellular uptake were evaluated in breast cancer cells. The particle size and zeta potential of the DTX-NPs were 160.5 ± 3.0 nm and -26.7 ± 0.46 mV, respectively. The encapsulation efficiency and drug loading were 81.3 ± 1.85% and 10.6 ± 0.24%, respectively. The in vitro release of DTX from the DTX-NPs was sustained at pH 7.4 containing 0.5% Tween 80. The viability of MDA-MB-231 and MCF-7 cells with DTX-NPs was 37.5 ± 0.5% and 30.3 ± 1.13%, respectively. The IC50 values of DTX-NPs were 3.92- and 6.75-fold lower than that of DTX for MDA-MB-231 cells and MCF-7 cells, respectively. The cellular uptake of coumarin-6-loaded PLGA-NPs in MCF-7 cells was significantly higher than that in MDA-MB-231 cells. The pharmacological sensitivity in breast cancer cells was higher on MCF-7 cells than on MDA-MB-231 cells. In conclusion, we successfully developed DTX-NPs that showed a great potential for the controlled release of DTX. DTX-NPs are an effective formulation for improving anticancer effect in breast cancer cells.

키워드

과제정보

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (NRF-2020R1F1A1075127 and NRF-2021R1A2C1011176).

참고문헌

  1. Lovitt CJ, Shelper TB, Avery VM. Doxorubicin resistance in breast cancer cells is mediated by extracellular matrix proteins. BMC Cancer. 2018;18:41. https://doi.org/10.1186/s12885-017-3953-6
  2. Xie Z, Zhang Y, Jin C, Fu D. Gemcitabine-based chemotherapy as a viable option for treatment of advanced breast cancer patients: a meta-analysis and literature review. Oncotarget. 2017;9:7148-7161. https://doi.org/10.18632/oncotarget.23426
  3. Martin M. nab-Paclitaxel dose and schedule in breast cancer. Breast Cancer Res. 2015;17:81. https://doi.org/10.1186/s13058-015-0587-y
  4. Sorolla A, Wang E, Clemons TD, Evans CW, Plani-Lam JH, Golden E, Dessauvagie B, Redfern AD, Swaminathan-Iyer K, Blancafort P. Triple-hit therapeutic approach for triple negative breast cancers using docetaxel nano-particles, EN1-iPeps and RGD peptides. Nanomedicine. 2019;20:102003. https://doi.org/10.1016/j.nano.2019.04.006
  5. Kendzierski DC, Schneider BP, Kiel PJ. Efficacy of different leuprolide administration schedules in premenopausal breast cancer: a retrospective review. Clin Breast Cancer. 2018;18:e939-e942.
  6. U.S. Food and Drug Administration. MedWatch: The FDA safety information and adverse event reporting program [Internet]. Silver Spring (MD): U.S. Food and Drug Administration, 1996 [cited 2020 May 12]. Available from: www.fda.gov/medwatch.
  7. U.S. Food and Drug Administration. Taxotere® 2015 [Internet]. Silver Spring (MD): U.S. Food and Drug Administration, 2015 [cited 2020 Jul 17]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/020449orig1s075.pdf.
  8. Gaucher G, Marchessault RH, Leroux JC. Polyester-based micelles and nanoparticles for the parenteral delivery of taxanes. J Control Release. 2010;143:2-12. https://doi.org/10.1016/j.jconrel.2009.11.012
  9. Engels FK, Mathot RA, Verweij J. Alternative drug formulations of docetaxel: a review. Anticancer Drugs. 2007;18:95-103. https://doi.org/10.1097/CAD.0b013e3280113338
  10. Mi Y, Guo Y, Feng SS. Nanomedicine for multimodality treatment of cancer. Nanomedicine (Lond). 2012;7:1791-1794. https://doi.org/10.2217/nnm.12.159
  11. Kutty RV, Feng SS. Cetuximab conjugated vitamin E TPGS micelles for targeted delivery of docetaxel for treatment of triple negative breast cancers. Biomaterials. 2013;34:10160-10171. https://doi.org/10.1016/j.biomaterials.2013.09.043
  12. Makadia HK, Siegel SJ. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers (Basel). 2011;3:1377-1397. https://doi.org/10.3390/polym3031377
  13. Blasi P. Poly(lactic acid)/poly(lactic-co-glycolic acid)-based microparticles: an overview. J Pharm Investig. 2019;49:337-346. https://doi.org/10.1007/s40005-019-00453-z
  14. Elmowafy EM, Tiboni M, Soliman ME. Biocompatibility, biodegradation and biomedical applications of poly(lactic acid)/poly(lacticco-glycolic acid) micro and nanoparticles. J Pharm Investig. 2019;49:347-380. https://doi.org/10.1007/s40005-019-00439-x
  15. Choi JS, Jang WS, Park JS. Comparison of adsorption and conjugation of Herceptin on poly(lactic-co-glycolic acid) nanoparticles - effect on cell internalization in breast cancer cells. Mater Sci Eng C Mater Biol Appl. 2018;92:496-507. https://doi.org/10.1016/j.msec.2018.06.059
  16. Gao J, Liu J, Xie F, Lu Y, Yin C, Shen X. Co-delivery of docetaxel and salinomycin to target both breast cancer cells and stem cells by PLGA/TPGS nanoparticles. Int J Nanomedicine. 2019;14:9199-9216. https://doi.org/10.2147/IJN.S230376
  17. Prabhuraj RS, Bomb K, Srivastava R, Bandyopadhyaya R. Selection of superior targeting ligands using PEGylated PLGA nanoparticles for delivery of curcumin in the treatment of triple-negative breast cancer cells. J Drug Deliv Sci Technol. 2020;57:101722. https://doi.org/10.1016/j.jddst.2020.101722
  18. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Preat V. PLGA-based nanoparticles: an overview of biomedical applications. J Control Release. 2012;161:505-522. https://doi.org/10.1016/j.jconrel.2012.01.043
  19. Berthet M, Gauthier Y, Lacroix C, Verrier B, Monge C. Nanoparticle-based dressing: the future of wound treatment? Trends Biotechnol. 2017;35:770-784. https://doi.org/10.1016/j.tibtech.2017.05.005
  20. Chambon P, Chen L, Furzeland S, Atkins D, Weaver JVM, Adams DJ. Poly(N-isopropylacrylamide) branched polymernanoparticles. Polym Chem. 2011;2:941-949. https://doi.org/10.1039/c0py00369g
  21. Perinelli DR, Cespi M, Bonacucina G, Palmieri GF. PEGylated polylactide (PLA) and poly (lactic-co-glycolic acid) (PLGA) copolymers for the design of drug delivery systems. J Pharm Investig. 2019;49:443-458. https://doi.org/10.1007/s40005-019-00442-2
  22. Zhang Y, Hou J, Qian C, Song L, Guo S. Taxol-loaded nanoparticles with methoxy poly(ethylene glycol)-b-poly(ε-caprolactone) as a novel additive in the outer aqueous phase. J Appl Polym Sci. 2011;121:2386-2393. https://doi.org/10.1002/app.33518
  23. Akhtar N, Ahad A, Khar RK, Jaggi M, Aqil M, Iqbal Z, Ahmad FJ, Talegaonkar S. The emerging role of P-glycoprotein inhibitors in drug delivery: a patent review. Expert Opin Ther Pat. 2011;21:561-576. https://doi.org/10.1517/13543776.2011.561784
  24. Mu L, Feng SS. Vitamin E TPGS used as emulsifier in the solvent evaporation/extraction technique for fabrication of polymeric nano-spheres for controlled release of paclitaxel (Taxol). J Control Release. 2002;80:129-144. https://doi.org/10.1016/S0168-3659(02)00025-1
  25. Guo Y, Luo J, Tan S, Otieno BO, Zhang Z. The applications of Vitamin E TPGS in drug delivery. Eur J Pharm Sci. 2013;49:175-186. https://doi.org/10.1016/j.ejps.2013.02.006
  26. Wang G, Yu B, Wu Y, Huang B, Yuan Y, Liu CS. Controlled preparation and antitumor efficacy of vitamin E TPGS-functionalized PLGA nanoparticles for delivery of paclitaxel. Int J Pharm. 2013;446:24-33. https://doi.org/10.1016/j.ijpharm.2013.02.004
  27. Zhang Z, Lee SH, Gan CW, Feng SS. In vitro and in vivo investigation on PLA-TPGS nanoparticles for controlled and sustained small molecule chemotherapy. Pharm Res. 2008;25:1925-1935. https://doi.org/10.1007/s11095-008-9611-6
  28. Shieh MJ, Hsu CY, Huang LY, Chen HY, Huang FH, Lai PS. Reversal of doxorubicin-resistance by multifunctional nanoparticles in MCF-7/ADR cells. J Control Release. 2011;152:418-425. https://doi.org/10.1016/j.jconrel.2011.03.017
  29. Yang C, Wu T, Qi Y, Zhang Z. Recent advances in the application of vitamin E TPGS for drug delivery. Theranostics. 2018;8:464-485. https://doi.org/10.7150/thno.22711
  30. Neuzil J, Tomasetti M, Mellick AS, Alleva R, Salvatore BA, Birringer M, Fariss MW. Vitamin E analogues: a new class of inducers of apoptosis with selective anti-cancer effects. Curr Cancer Drug Targets. 2004;4:355-372. https://doi.org/10.2174/1568009043332943
  31. Win KY, Feng SS. Effects of particle size and surface coating on cellular uptake of polymeric nanoparticles for oral delivery of anticancer drugs. Biomaterials. 2005;26:2713-2722. https://doi.org/10.1016/j.biomaterials.2004.07.050
  32. Gan CW, Feng SS. Transferrin-conjugated nanoparticles of poly(lactide)-D-alpha-tocopheryl polyethylene glycol succinate diblock copolymer for targeted drug delivery across the blood-brain barrier. Biomaterials. 2010;31:7748-7757. https://doi.org/10.1016/j.biomaterials.2010.06.053
  33. Zhang Z, Feng SS. Nanoparticles of poly(lactide)/vitamin E TPGS copolymer for cancer chemotherapy: synthesis, formulation, characterization and in vitro drug release. Biomaterials. 2006;27:262-270. https://doi.org/10.1016/j.biomaterials.2005.05.104
  34. Varma MV, Panchagnula R. Enhanced oral paclitaxel absorption with vitamin E-TPGS: effect on solubility and permeability in vitro, in situ and in vivo. Eur J Pharm Sci. 2005;25:445-453. https://doi.org/10.1016/j.ejps.2005.04.003
  35. Noori Koopaei M, Khoshayand MR, Mostafavi SH, Amini M, Khorramizadeh MR, Jeddi Tehrani M, Atyabi F, Dinarvand R. Docetaxel loaded PEG-PLGA nanoparticles: optimized drug loading, in-vitro cytotoxicity and in-vivo antitumor effect. Iran J Pharm Res. 2014;13:819-833.
  36. Choi JS, Kwon SH, Lee SE, Jang WS, Byeon JC, Jeong HM, Park JS. Use of acidifier and solubilizer in tadalafil solid dispersion to enhance the in vitro dissolution and oral bioavailability in rats. Int J Pharm. 2017;526:77-87. https://doi.org/10.1016/j.ijpharm.2017.04.056
  37. Li A, Zuo L. Construction of anti-EpCAM drug-loaded immunomagnetic balls and its application in diagnosis of breast cancer. Nano Life. 2019;9:1940006. https://doi.org/10.1142/S1793984419400063
  38. Kulhari H, Pooja D, Shrivastava S, Telukutala SR, Barui AK, Patra CR, Naidu Vegi GM, Adams DJ, Sistla R. Cyclic-RGDfK peptide conjugated succinoyl-TPGS nanomicelles for targeted delivery of docetaxel to integrin receptor over-expressing angiogenic tumours. Nanomedicine. 2015;11:1511-1520. https://doi.org/10.1016/j.nano.2015.04.007
  39. Jiang P, Yu D, Zhang W, Mao Z, Gao C. Influence of bovine serum albumin coated poly(lactic-co-glycolic acid) particles on differentiation of mesenchymal stem cells. RSC Adv. 2015;5:40924-40931. https://doi.org/10.1039/C5RA07219K
  40. Wang Y, Li P, Kong L. Chitosan-modified PLGA nanoparticles with versatile surface for improved drug delivery. AAPS PharmSciTech. 2013;14:585-592. https://doi.org/10.1208/s12249-013-9943-3
  41. de Souza Pietra RCC, Cruz RC, Melo CN, Rodrigues LB, Santos PC, Bretz GPM, Soares BM, de Sousa GR, Ferreira MVL, Cisalpino PS, Magalhaes PP, de Macedo Farias L, Pinotti M. Evaluation of polymeric PLGA nanoparticles conjugated to curcumin for use in aPDT. Braz J Pharm Sci. 2017;53:e16043.
  42. Jang MH, Kim CH, Yoon HY, Sung SW, Goh MS, Lee ES, Shin DJ, Choi YW. Steric stabilization of RIPL peptide-conjugated liposomes and in vitro assessment. J Pharm Investig. 2019;49:115-125. https://doi.org/10.1007/s40005-018-0392-6
  43. Choi JS, Park JS. Design and evaluation of the anticancer activity of paclitaxel-loaded anisotropic-poly(lactic-co-glycolic acid) nanoparticles with PEGylated chitosan surface modifications. Int J Biol Macromol. 2020;162:1064-1075. https://doi.org/10.1016/j.ijbiomac.2020.06.237
  44. Sharma N, Madan P, Lin S. Effect of process and formulation variables on the preparation of parenteral paclitaxel-loaded biodegradable polymeric nanoparticles: a co-surfactant study. Asian J Pharm Sci. 2016;11:404-416. https://doi.org/10.1016/j.ajps.2015.09.004
  45. Krishnamachari Y, Madan P, Lin S. Development of pH- and time-dependent oral microparticles to optimize budesonide delivery to ileum and colon. Int J Pharm. 2007;338:238-247. https://doi.org/10.1016/j.ijpharm.2007.02.015
  46. Gabor F, Ertl B, Wirth M, Mallinger R. Ketoprofen-poly(D,L-lacticco-glycolic acid) microspheres: influence of manufacturing parameters and type of polymer on the release characteristics. J Microencapsul. 1999;16:1-12. https://doi.org/10.1080/026520499289266
  47. Rafiei P, Haddadi A. Docetaxel-loaded PLGA and PLGA-PEG nanoparticles for intravenous application: pharmacokinetics and biodistribution profile. Int J Nanomedicine. 2017;12:935-947. https://doi.org/10.2147/IJN.S121881
  48. Qureshi OS, Kim HS, Zeb A, Choi JS, Kim HS, Kwon JE, Kim MS, Kang JH, Ryou C, Park JS, Kim JK. Sustained release docetaxel-incorporated lipid nanoparticles with improved pharmacokinetics for oral and parenteral administration. J Microencapsul. 2017;34:250-261. https://doi.org/10.1080/02652048.2017.1337247