CC BY 4.0 · Pharmaceutical Fronts 2022; 04(03): e188-e196
DOI: 10.1055/s-0042-1757511
Original Article

Novel Docetaxel-Loaded Micelles Based on all-trans-Retinoic Acid: Preparation and Pharmacokinetic Study in Rats

Ya-Ni Yang#
1   National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
,
Jia-Jia Cheng#
1   National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
,
Jun He
1   National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
,
Wei-Gen Lu
1   National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
› Author Affiliations

Abstract

Docetaxel (DTX) is a poorly soluble drug. The purpose of this study was to explore a DTX-loaded micelle delivery system using N-(all-trans-retinoyl)-L-cysteic acid methyl ester sodium salt (XMeNa) as the carrier materials. In this study, amphiphilic surfactant XMeNa was synthesized. Then, the blood biocompatibility and the value of critical micelle concentration (CMC) were assessed by a hemolysis test and pyrene-based fluorescent probe techniques, respectively. The XM-DTX micelles were prepared using the method of thin-film hydration, and characterized by dynamic light scattering and transmission electron microscopy (TEM). The entrapment efficiency (EE) and drug loading efficiency (DLE) were assessed by the ultrafiltration method. In vitro release and pharmacokinetic behaviors of XM-DTX micelles were performed in rats using Taxotere (a commercialized DTX injection) as a control. Our data confirmed the excellent blood biocompatibility of XMeNa as a carrier. XMeNa can self-assemble into micelles in aqueous media with a very low CMC (6.2 μg/mL). The average size and zeta potential of the XM-DTX micelles were 17.3 ± 0.2 nm, and −41.6 ± 0.3 mV, respectively. EE and DLE reached up to 95.3 ± 0.7% and 22.4 ± 0.2%, respectively, which may account for the high solubility of DTX in normal saline. The micelles were spherical in TEM with good dispersion and no aggregation and adhesion, and exhibited good stability after reconstitution over 8 hours. Results from in vitro release assay suggested a much slower release behavior of XM-DTX micelles in comparison to Taxotere. Additionally, XM-DTX micelles prolonged DTX retention in blood circulation, increased the area under the curve by 2.4-fold, and significantly decreased the clearance of the drug. Given above, the XM-DTX micelles could improve the solubility and the release of DTX. The amphiphilic surfactant XMeNa also exhibited great potential as a vehicle for exploring delivery of poorly water soluble drugs in the future.

# These authors contributed equally to this work.




Publication History

Received: 17 April 2022

Accepted: 16 June 2022

Article published online:
19 September 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Ganju A, Yallapu MM, Khan S, Behrman SW, Chauhan SC, Jaggi M. Nanoways to overcome docetaxel resistance in prostate cancer. Drug Resist Updat 2014; 17 (1–2): 13-23
  • 2 Martin M, Pienkowski T, Mackey J. et al. Breast Cancer International Research Group 001 Investigators. Adjuvant docetaxel for node-positive breast cancer. N Engl J Med 2005; 352 (22) 2302-2313
  • 3 A. Razak SA, Mohd Gazzali A, Fisol FA. et al. Advances in nanocarriers for effective delivery of docetaxel in the treatment of lung cancer: an overview. Cancers (Basel) 2021; 13 (03) 400
  • 4 Imran M, Saleem S, Chaudhuri A. et al. Docetaxel: an update on its molecular mechanisms, therapeutic trajectory and nanotechnology in the treatment of breast, lung and prostate cancer. J Drug Deliv Sci Technol 2020; 60: 101959
  • 5 Sohail MF, Rehman M, Sarwar HS. et al. Advancements in the oral delivery of Docetaxel: challenges, current state-of-the-art and future trends. Int J Nanomedicine 2018; 13: 3145-3161
  • 6 Engels FK, Mathot RA, Verweij J. Alternative drug formulations of docetaxel: a review. Anticancer Drugs 2007; 18 (02) 95-103
  • 7 Schwartzberg LS, Navari RM. Safety of polysorbate 80 in the oncology setting. Adv Ther 2018; 35 (06) 754-767
  • 8 Zhang L, Zhang N. How nanotechnology can enhance docetaxel therapy. Int J Nanomedicine 2013; 8: 2927-2941
  • 9 Ghamkhari A, Pouyafar A, Salehi R, Rahbarghazi R. Chrysin and docetaxel loaded biodegradable micelle for combination chemotherapy of cancer stem cell. Pharm Res 2019; 36 (12) 165
  • 10 Lee SW, Yun MH, Jeong SW. et al. Development of docetaxel-loaded intravenous formulation, Nanoxel-PM™ using polymer-based delivery system. J Control Release 2011; 155 (02) 262-271
  • 11 Hu Q, Rijcken CJ, Bansal R, Hennink WE, Storm G, Prakash J. Complete regression of breast tumour with a single dose of docetaxel-entrapped core-cross-linked polymeric micelles. Biomaterials 2015; 53: 370-378
  • 12 Atrafi F, Dumez H, Mathijssen RHJ. et al. A phase I dose-escalation and pharmacokinetic study of a micellar nanoparticle with entrapped docetaxel (CPC634) in patients with advanced solid tumours. J Control Release 2020; 325: 191-197
  • 13 Zhang Y, Huang Y, Li S. Polymeric micelles: nanocarriers for cancer-targeted drug delivery. AAPS PharmSciTech 2014; 15 (04) 862-871
  • 14 Lu Y, Zhang E, Yang J, Cao Z. Strategies to improve micelle stability for drug delivery. Nano Res 2018; 11 (10) 4985-4998
  • 15 Cheng M, Liu QM, Liu W. et al. Engineering micelles for the treatment and diagnosis of atherosclerosis. J Drug Deliv Sci Technol 2021; 63: 102473
  • 16 Zheng X, Xie JZ, Zhang X. et al. An overview of polymeric nanomicelles in clinical trials and on the market. Chin Chem Lett 2021; 32: 243-257
  • 17 European Medicines Agency. Assessment report for Apealea, EMA/CHMP/785964/2018. 2018;1–121. Accessed February 25, 2022 at: https://www.ema.europa.eu/en/documents/assessment-report/apealea-epar-public-assessment-report_en.pdf.
  • 18 Vergote I, Bergfeldt K, Franquet A. et al. A randomized phase III trial in patients with recurrent platinum sensitive ovarian cancer comparing efficacy and safety of paclitaxel micellar and Cremophor EL-paclitaxel. Gynecol Oncol 2020; 156 (02) 293-300
  • 19 Borgå O, Lilienberg E, Bjermo H, Hansson F, Heldring N, Dediu R. Pharmacokinetics of total and unbound paclitaxel after administration of paclitaxel micellar or nab-paclitaxel: an open, randomized, cross-over, explorative study in breast cancer patients. Adv Ther 2019; 36 (10) 2825-2837
  • 20 Strelchenok O, Aleksov J. Retinol derivatives, their use in the treatment of cancer and for potentiating the efficacy of other cytotoxic agents. WO Patent 2002SE02087. November, 2002
  • 21 He J, Cheng JJ, Wang ZF. et al. Preparation method of sodium salt of N-(all trans-retinol)-L-cystathionine methyl ester. CN Patent 110218168A. September, 2019
  • 22 Aguiar J, Carpena P, Molina-Bolívar JA. et al. On the determination of the critical micelle concentration by the pyrene 1:3 ratio method. J Colloid Interface Sci 2003; 258: 116-122
  • 23 Fu PP, Cheng SH, Coop L. et al. Photoreaction, phototoxicity, and photocarcinogenicity of retinoids. J Environ Sci Health Part C Environ Carcinog Ecotoxicol Rev 2003; 21 (02) 165-197
  • 24 Kore G, Kolate A, Nej A, Misra A. Polymeric micelle as multifunctional pharmaceutical carriers. J Nanosci Nanotechnol 2014; 14 (01) 288-307
  • 25 Torchilin V. Tumor delivery of macromolecular drugs based on the EPR effect. Adv Drug Deliv Rev 2011; 63 (03) 131-135
  • 26 Patil S, Sandberg A, Heckert E, Self W, Seal S. Protein adsorption and cellular uptake of cerium oxide nanoparticles as a function of zeta potential. Biomaterials 2007; 28 (31) 4600-4607
  • 27 Alexis F, Pridgen E, Molnar LK, Farokhzad OC. Factors affecting the clearance and biodistribution of polymeric nanoparticles. Mol Pharm 2008; 5 (04) 505-515
  • 28 Abouelmagd SA, Sun B, Chang AC, Ku YJ, Yeo Y. Release kinetics study of poorly water-soluble drugs from nanoparticles: are we doing it right?. Mol Pharm 2015; 12 (03) 997-1003
  • 29 Lee JH, Yeo Y. Controlled drug release from pharmaceutical nanocarriers. Chem Eng Sci 2015; 125: 75-84
  • 30 Kang M, Lee B, Leal C. Three-dimensional microphase separation and synergistic permeability in stacked lipid-polymer hybrid membranes. Chem Mater 2017; 29 (21) 9120-9132
  • 31 Zoya I, He HS, Wang LT. et al. The intragastrointestinal fate of paclitaxel-loaded micelles: implications on oral drug delivery. Chin Chem Lett 2021; 32: 1545-1549
  • 32 Zhao J, Chai YD, Zhang J, Huang PF, Nakashima K, Gong YK. Long circulating micelles of an amphiphilic random copolymer bearing cell outer membrane phosphorylcholine zwitterions. Acta Biomater 2015; 16: 94-102
  • 33 Zheng P, Liu Y, Chen JJ. et al. Targeted pH-responsive polyion complex micelle for controlled intracellular drug delivery. Chin Chem Lett 2020; 31: 1178-1182