Generic placeholder image

Current Nanomedicine

Editor-in-Chief

ISSN (Print): 2468-1873
ISSN (Online): 2468-1881

Research Article

Kinetic Evaluation of Anti-tumor Chlorambucil Release from O-stearoyl Mannose PLGA Nanoparticles

Author(s): Antonio O. Costa, Claure N. Lunardi and Anderson J. Gomes*

Volume 10, Issue 1, 2020

Page: [63 - 75] Pages: 13

DOI: 10.2174/2468187309666190823153341

Abstract

Purpose: This study assesses the kinetics of the anti-tumor drug chlorambucil (CLB) incorporated into PLGA nanoparticles (NP-CLB) with and without the presence of the O-stearoyl mannose (OEM) functionalizing agent (NP-CLBMAN).

Methods: OEM was synthesized and used in the NP-CLB-MAN formulation. The nanoparticles were characterized by dynamic light scattering, electrophoretic light scattering, scanning electron microscopy, and Fourier-transform infrared spectroscopy.

Results: The nanoparticles presented an encapsulation efficiency greater than 61% and a PdI between 0.186–0.217. The mean size was 185 nm for NP-CLB and 220 nm for NPCLB- MAN, and the zeta potential values were -17.7 mV for NP-CLB and -14.2 mV for NP- CLB-MAN. Scanning electron microscopy showed that NPs with OEM have a surface with a different shape, and FTIR analyses showed binding of CLB to the drug delivery system, as well as functionalization with OEM. In vitro release studies showed a biphasic release profile for both systems, and they were analyzed considering the mathematical Korsmeyer-Peppas, first-order, and Fick diffusion models, and the combination of the first-order and Fick diffusion models.

Conclusion: The experimental results obtained for the release of CLB were better described using a combination of the first order and Fick diffusion mathematical models.

Keywords: PLGA, nanoparticles, o-stearoyl mannose, chlorambucil, mathematical modeling.

Graphical Abstract
[1]
Tiwari G, Tiwari R, Sriwastawa B, et al. Drug delivery systems: An updated review. Int J Pharm Investig 2012; 2(1): 2-11.
[http://dx.doi.org/10.4103/2230-973X.96920] [PMID: 23071954]
[2]
Sadat Tabatabaei Mirakabad F, Nejati-Koshki K, Akbarzadeh A, et al. PLGA-based nanoparticles as cancer drug delivery systems. Asian Pac J Cancer Prev 2014; 15(2): 517-35.
[http://dx.doi.org/10.7314/APJCP.2014.15.2.517] [PMID: 24568455]
[3]
Makadia HK, Siegel SJ. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers (Basel) 2011; 3(3): 1377-97.
[http://dx.doi.org/10.3390/polym3031377] [PMID: 22577513]
[4]
Bala I, Hariharan S, Kumar MN. PLGA nanoparticles in drug delivery: the state of the art. Crit Rev Ther Drug Carrier Syst 2004; 21(5): 387-422.
[http://dx.doi.org/10.1615/CritRevTherDrugCarrierSyst.v21.i5.20]
[5]
Iyer AK, Khaled G, Fang J, Maeda H. Exploiting the enhanced permeability and retention effect for tumor targeting. Drug Discov Today 2006; 11(17-18): 812-8.
[http://dx.doi.org/10.1016/j.drudis.2006.07.005] [PMID: 16935749]
[6]
Chittasupho C, Xie SX, Baoum A, Yakovleva T, Siahaan TJ, Berkland CJ. ICAM-1 targeting of doxorubicin-loaded PLGA nanoparticles to lung epithelial cells. Eur J Pharm Sci 2009; 37(2): 141-50.
[http://dx.doi.org/10.1016/j.ejps.2009.02.008] [PMID: 19429421]
[7]
Mahapatro A, Singh DK. Biodegradable nanoparticles are excellent vehicle for site directed in-vivo delivery of drugs and vaccines. J Nanobiotechnology 2011; 9(1): 55.
[http://dx.doi.org/10.1186/1477-3155-9-55] [PMID: 22123084]
[8]
Everett JL, Roberts JJ, Ross WCJ. 486 Aryl-2-halogenoalkylamines Part XII Some carboxylic derivatives of NN-di-2-chloroethylaniline J Chem Soc (Resumed) 1953. 0: 2386-92.
[9]
Ji X, Shi C, Li N, Wang K, Li Z, Luan Y. Catanionic drug-derivative nano-objects constructed by chlorambucil and its derivative for efficient leukaemia therapy. Colloids Surf B Biointerfaces 2015; 136: 1081-8.
[http://dx.doi.org/10.1016/j.colsurfb.2015.11.016] [PMID: 26595388]
[10]
Dias DJ, Joanitti GA, Azevedo RB, Silva LP, Lunardi CN, Gomes AJ. Chlorambucil encapsulation into PLGA nanoparticles and cytotoxic effects in breast cancer cell. J Biophys Chem 2015; 6(1): 1-6.
[http://dx.doi.org/10.4236/jbpc.2015.61001]
[11]
Fan M, Liang X, Li Z, Wang H, Yang D, Shi B. Chlorambucil gemcitabine conjugate nanomedicine for cancer therapy. Eur J Pharm Sci 2015; 79: 20-6.
[http://dx.doi.org/10.1016/j.ejps.2015.08.013] [PMID: 26342774]
[12]
Horie K. Definitions of terms relating to reactions of polymers and to functional poly- meric materials (IUPAC Recommendations 2003). Pure Appl Chem 2004; 76(4): 889-906.
[http://dx.doi.org/10.1351/pac200476040889]
[13]
Byrne JD, Betancourt T, Brannon-Peppas L. Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev 2008; 60(15): 1615-26.
[http://dx.doi.org/10.1016/j.addr.2008.08.005] [PMID: 18840489]
[14]
Ghosh S, Das S, De AK, Kar N, Bera T. Amphotericin B- loaded mannose modified poly (D, L-lactide-co-glycolide) polymeric nanoparticles for the treatment of visceral leish-maniasis: in vitro and in vivo approaches. RSC Advances 2017; 7(47): 29575-90.
[http://dx.doi.org/10.1039/C7RA04951J]
[15]
Zhu S, Niu M, O’Mary H, Cui Z. Targeting of tumor-associated macrophages made possible by PEG-sheddable, mannose-modified nanoparticles. Mol Pharm 2013; 10(9): 3525-30.
[http://dx.doi.org/10.1021/mp400216r] [PMID: 23901887]
[16]
Halamoda-Kenzaoui B, Bremer-Hoffmann S. Main trends of immune effects triggered by nanomedicines in preclinical studies. Int J Nanomedicine 2018; 13: 5419-31.
[http://dx.doi.org/10.2147/IJN.S168808] [PMID: 30271138]
[17]
Moghimi SM, Simberg D. Complement activation turnover on surfaces of nanoparticles. Nano Today 2017; 15: 8-10.
[http://dx.doi.org/10.1016/j.nantod.2017.03.001] [PMID: 29399037]
[18]
Dobrovolskaia MA, Aggarwal P, Hall JB, McNeil SE. Preclinical studies to understand nanoparticle interaction with the immune system and its potential effects on nanoparticle biodistribution. Mol Pharm 2008; 5(4): 487-95.
[http://dx.doi.org/10.1021/mp800032f] [PMID: 18510338]
[19]
Stepien G, Moros M, Pérez-Hernández M, et al. Effect of surface chemistry and associated protein corona on the long-term biodegradation of iron oxide nanopar- ticles in vivo. ACS Appl Mater Interfaces 2018; 10(5): 4548-60.
[20]
Wei Y, Quan L, Zhou C, Zhan Q. Factors relating to the biodistribution & clearance of nanoparticles & their effects on in vivo application. Nanomedicine (Lond) 2018; 13(12): 1495-512.
[http://dx.doi.org/10.2217/nnm-2018-0040] [PMID: 29972677]
[21]
Croissant JG, Fatieiev Y, Julfakyan K, et al. Biodegradable oxamide-phenylene-based mesoporous organosilica nanoparticles with unprecedented drug payloads for delivery in cells. Chemistry 2016; 22(42): 14806-11.
[http://dx.doi.org/10.1002/chem.201601714] [PMID: 27258139]
[22]
Gao H, He Q. The interaction of nanoparticles with plasma proteins and the consequent influence on nanoparticles behavior. Expert Opin Drug Deliv 2014; 11(3): 409-20.
[http://dx.doi.org/10.1517/17425247.2014.877442] [PMID: 24397260]
[23]
Duan X, Li Y. Physicochemical characteristics of nanoparticles affect circulation, biodistribution, cellular internalization, and trafficking. Small 2013; 9(9-10): 1521-32.
[http://dx.doi.org/10.1002/smll.201201390] [PMID: 23019091]
[24]
Chow EKH, Ho D. Cancer nanomedicine: from drug delivery to imaging. Sci Transl Med 2013; 5(216) 216rv4
[http://dx.doi.org/10.1126/scitranslmed.3005872]
[25]
Costa P, Sousa Lobo JM. Modeling and comparison of dissolution profiles. Eur J Pharm Sci 2001; 13(2): 123-33.
[http://dx.doi.org/10.1016/S0928-0987(01)00095-1] [PMID: 11297896]
[26]
Petralito S, Zanardi I, Memoli A, Annesini MC, Millucci V, Travagli V, Eds. Apparent solubility and dissolution profile at non-sink conditions as quality improvement. 2012.
[27]
Gutiérrez-Valenzuela CA, Esquivel R, Guerrero-Germán P, et al. Evaluation of a combined emulsion process to en- capsulate methylene blue into PLGA nanoparticles. RSC Advances 2018; 8(1): 414-22.
[http://dx.doi.org/10.1039/C7RA12296A]
[28]
Lucero-Acuña A, Guzmán R. Nanoparticle encapsulation and controlled release of a hydrophobic kinase inhibitor: Three stage mathematical modeling and parametric analysis. Int J Pharm 2015; 494(1): 249-57.
[http://dx.doi.org/10.1016/j.ijpharm.2015.07.049] [PMID: 26216413]
[29]
Manadas R, Pina ME, Veiga F. A dissolução in vitro na previsão da absorção oral de fármacos em formas farma- cêuticas de liberação modificada. Revista Brasileira de Ciências Farmacêuticas 2002; 38(4): 375-99.
[30]
Lopes CM, Lobo JMS, Costa P. Formas farmacêuticas de liberação modificada: polímeros hidrifílicos. Revista Brasileira de Ciências Farmacêuticas 2005; 41(2): 143-54.
[http://dx.doi.org/10.1590/S151693322005000200003]
[31]
Ritge PL, Peppas NA. A simple equation for descrip- tion of solute release II. Fickian and anomalous release from swellable devices. J Control Release 1987; 5(1): 37-42.
[http://dx.doi.org/10.1016/0168-3659(87)90035-6]
[32]
Dash S, Murthy PN, Nath L, Chowdhury P. Kinetic modeling on drug release from controlled drug delivery systems. Acta Pol Pharm 2010; 67(3): 217-23.
[PMID: 20524422]
[33]
Crank J. The mathematics of diffusion. 2nd ed. London: Oxford University Press 1975.
[34]
Betts JG, Desaix P, Johnson E, et al. Anatomy and physiology. Texas: OpenStax - Rice University 2013.
[35]
Acharya S, Sahoo SK. PLGA nanoparticles containing various anticancer agents and tumour delivery by EPR effect. Adv Drug Deliv Rev 2011; 63(3): 170-83.
[http://dx.doi.org/10.1016/j.addr.2010.10.008] [PMID: 20965219]
[36]
Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol 2015; 33(9): 941-51.
[http://dx.doi.org/10.1038/nbt.3330] [PMID: 26348965]
[37]
Tavano R, Segat D, Reddi E, et al. Procoagulant properties of bare and highly PEGylated vinyl-modified silica nanoparticles. Nanomedicine (Lond) 2010; 5(6): 881-96.
[http://dx.doi.org/10.2217/nnm.10.65] [PMID: 20735224]
[38]
Ilinskaya AN, Dobrovolskaia MA. Nanoparticles and the blood coagulation system 2016.
[http://dx.doi.org/10.1142/9789813140455_0008]
[39]
Silverstein RM, Webster FX, Kiemle DJ. Spectrometric identification of organic compounds. 7th ed. Hoboken: John Siley & Sons 2005.
[40]
Lunardi CN, Gomes AJ, Palepu S, Galwaduge PT, Hillman EM. PLGA nano/microparticles loaded with cresyl violet as a tracer for drug delivery: Characterization and in-situ hyperspectral fluorescence and 2-photon localization. Mater Sci Eng C 2017; 70(Pt 1): 505-11.
[http://dx.doi.org/10.1016/j.msec.2016.09.020] [PMID: 27770922]
[41]
Messaritaki A, Black SJ, van der Walle CF, Rigby SP. NMR and confocal microscopy studies of the mechanisms of burst drug release from PLGA microspheres. J Control Release 2005; 108(2-3): 271-81.
[http://dx.doi.org/10.1016/j.jconrel.2005.08.010] [PMID: 16169112]
[42]
Senapati S, Thakur R, Verma SP, et al. Layered double hydroxides as effective carrier for anticancer drugs and tailoring of release rate through interlayer anions. J Control Release 2016; 224: 186-98.
[http://dx.doi.org/10.1016/j.jconrel.2016.01.016] [PMID: 26774219]

© 2024 Bentham Science Publishers | Privacy Policy