Generic placeholder image

Recent Patents on Drug Delivery & Formulation

Editor-in-Chief

ISSN (Print): 1872-2113
ISSN (Online): 2212-4039

Review Article

Preparation and Surface Modification of Polymeric Nanoparticles for Drug Delivery: State of the Art

Author(s): Garima Joshi, Mitali Patel, Deepak Chaudhary and Krutika Sawant*

Volume 14, Issue 3, 2020

Page: [201 - 213] Pages: 13

DOI: 10.2174/1872211314666200904105036

Price: $65

Abstract

Nanotechnology is one of the emerging fields in drug delivery for targeting the drug to the site of action. The polymeric nanoparticles as drug delivery systems have gained importance for the last few decades. They offer advantages over liposomes, dendrimers, emulsions etc. Surface engineering of polymeric nanoparticles is widely utilized to effectively target the cells in various diseases such as cancer, HIV infection. Surface modified nanoparticles offer various advantages such as targeted drug delivery, reduction in side effects, dose reduction and improved therapeutic efficacy. Moreover, they can aid in improving physical and biochemical properties, pharmacokinetic and pharmacodynamic profiles of the drug. Surface modified polymeric nanoparticles can provide targeted delivery of drugs into specific cells, especially when targets are intracellularly localized. This approach of surface modification would be more advantageous for the delivery of various anticancer, anti-inflammatory, anti-HIV drugs for more effective therapy. This review focuses on the techniques used for the fabrication of polymeric nanoparticles, the material used for surface modification and their applications.

Keywords: Nanoparticles, surface modification, drug delivery, Poly(lactic-co-glycolicacid) PLGA, polymeric, ligand.

Graphical Abstract
[1]
Kumari A, Yadav SK, Yadav SC. Biodegradable polymeric nanoparticles based drug delivery systems. Colloids Surf B Biointerfaces 2010; 75(1): 1-18.
[http://dx.doi.org/10.1016/j.colsurfb.2009.09.001]
[2]
He C, Hu Y, Yin L, Tang C, Yin C. Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomat 2010; 31(13): 3657-66.
[http://dx.doi.org/10.1016/j.biomaterials.2010.01.065]
[3]
Aziz Z, Ahmad A, Mohd-Setapar SH, et al. Recent advances in drug delivery of polymeric nano-micelles. Curr Drug Metab 2017; 18(1): 16-29.
[http://dx.doi.org/10.2174/1389200217666160921143616]
[4]
Chuo SC, Ahmad A, Mohd-Setapar SH, Ripin A. Reverse micelle extraction-an alternative for recovering antibiotics. Pharma Chem 2014; 6: 37-44.
[5]
Ahmad A, Khatoon A, Mohd-Setapar SH, et al. Effect of parameter on forward extraction of amoxicillin by using mixed reverse micelles. Res J Biotechnol 2013; 8: 10-4.
[6]
Marin E, Briceño MI, Caballero-George C. Critical evaluation of biodegradable polymers used in nanodrugs. Int J Nanomedicine 2013; 8: 3071-90.
[7]
Kulkarni SA, Feng SS. Effects of particle size and surface modification on cellular uptake and biodistribution of polymeric nanoparticles for drug delivery. Pharm Res 2013; 30(10): 2512-22.
[http://dx.doi.org/10.1007/s11095-012-0958-3]
[8]
Rao JP, Geckeler KE. Polymer nanoparticles: Preparation techniques and size-control parameters. Prog Polym Sci 2011; 36(7): 887-913.
[http://dx.doi.org/10.1016/j.progpolymsci.2011.01.001]
[9]
Nagavarma BV, Yadav HK, Ayaz AV, Vasudha LS, Shivakumar HG. Different techniques for preparation of polymeric nanoparticles-a review. Asian J Pharm Clin Res 2012; 5(3): 16-23.
[10]
Perez C, Sanchez A, Putnam D, Ting D, Langer R, Alonso MJ. Poly(lactic acid)-poly(ethylene glycol) nanoparticles as new carriers for the delivery of plasmid DNA. J Control Release 2001; 75(1-2): 211-24.
[http://dx.doi.org/10.1016/S0168-3659(01)00397-2]
[11]
Lu W, Zhang Y, Tan YZ, Hu KL, Jiang XG, Fu SK. Cationic albumin-conjugated pegylated nanoparticles as novel drug carrier for brain delivery. J Control Release 2005; 107(3): 428-48.
[http://dx.doi.org/10.1016/j.jconrel.2005.03.027]
[12]
Saxena V, Sadoqi M, Shao J. Indocyanine green-loaded biodegradable nanoparticles: preparation, physicochemical characterization and in vitro release. Int J Pharm 2004; 278(2): 293-301.
[http://dx.doi.org/10.1016/j.ijpharm.2004.03.032]
[13]
El-Shabouri MH. Positively charged nanoparticles for improving the oral bioavailability of cyclosporin-A. Int J Pharm 2002; 249(1-2): 101-8.
[http://dx.doi.org/10.1016/S0378-5173(02)00461-1]
[14]
Vargas A, Pegaz B, Debefve E, et al. Improved photodynamic activity of porphyrin loaded into nanoparticles: an in vivo evaluation using chick embryos. Int J Pharm 2004; 286(1-2): 131-45.
[http://dx.doi.org/10.1016/j.ijpharm.2004.07.029]
[15]
Galindo-Rodriguez S, Allémann E, Fessi H, Doelker E. Physicochemical parameters associated with nanoparticle formation in the salting-out, emulsification-diffusion, and nanoprecipitation methods. Pharm Res 2004; 21(8): 1428-39.
[http://dx.doi.org/10.1023/B:PHAM.0000036917.75634.be]
[16]
York P. Strategies for particle design using supercritical fluid technologies. Pharm Sci Technol Today 1999; 2(11): 430-40.
[http://dx.doi.org/10.1016/S1461-5347(99)00209-6]
[17]
Liu M, Zhou Z, Wang X, et al. Formation of poly (L, D-lactide) spheres with controlled size by direct dialysis. Polymer (Guildf) 2007; 48(19): 5767-79.
[http://dx.doi.org/10.1016/j.polymer.2007.07.053]
[18]
Chronopoulou L, Fratoddi I, Palocci C, Venditti I, Russo MV. Osmosis based method drives the self-assembly of polymeric chains into micro- and nanostructures. Langmuir 2009; 25(19): 11940-6.
[http://dx.doi.org/10.1021/la9016382]
[19]
Bettencourt A, Almeida AJ. Poly (methyl methacrylate) particulate carriers in drug delivery. J Microencapsul 2012; 29(4): 353-67.
[http://dx.doi.org/10.3109/02652048.2011.651500]
[20]
Sahu M, Satapathy T, Bahadur S, et al. Preparation methods for nanoparticle: a smart carrier system for treatment of cancer. World J Pharm Res 2018; 7(9): 216-26.
[21]
Ekman B, Sjöholm I. Improved stability of proteins immobilized in microparticles prepared by a modified emulsion polymerization technique. J Pharm Sci 1978; 67(5): 693-6.
[http://dx.doi.org/10.1002/jps.2600670533]
[22]
Ham HT, Choi YS, Chee MG, Chung IJ. Singlewall carbon nanotubes covered with polystyrene nanoparticles by in situ miniemulsion polymerization. J Polym Sci A Polym Chem 2006; 44(1): 573-84.
[http://dx.doi.org/10.1002/pola.21185]
[23]
Watnasirichaikul S, Davies NM, Rades T, Tucker IG. Preparation of biodegradable insulin nanocapsules from biocompatible microemulsions. Pharm Res 2000; 17(6): 684-9.
[http://dx.doi.org/10.1023/A:1007574030674]
[24]
Gaudin F, Sintes-Zydowicz N. Core–shell biocompatible polyurethane nanocapsules obtained by interfacial step polymerisation in miniemulsion. Colloids Surf A Physicochem Eng Asp 2008; 331(1-2): 133-42.
[http://dx.doi.org/10.1016/j.colsurfa.2008.07.028]
[25]
Braunecker WA, Matyjaszewski K. Controlled/living radical polymerization: Features, developments, and perspectives. Prog Polym Sci 2007; 32(1): 93-146.
[http://dx.doi.org/10.1016/j.progpolymsci.2006.11.002]
[26]
Calvo P, Remunan‐Lopez C, Vila‐Jato JL, Alonso MJ. Novel hydrophilic chitosan‐polyethylene oxide nanoparticles as protein carriers. J Appl Polym Sci 1997; 63(1): 125-32.
[http://dx.doi.org/10.1002/(SICI)1097-4628(19970103)63:1<125:AID-APP13>3.0.CO;2-4]
[27]
Bose RJ, Lee SH, Park H. Lipid-based surface engineering of PLGA nanoparticles for drug and gene delivery applications. Biomater Res 2016; 20(1): 34.
[http://dx.doi.org/10.1186/s40824-016-0081-3]
[28]
Pillai CK, Paul W, Sharma CP. Chitin and chitosan polymers: Chemistry, solubility and fiber formation. Prog Polym Sci 2009; 34(7): 641-78.
[http://dx.doi.org/10.1016/j.progpolymsci.2009.04.001]
[29]
Muxika A, Etxabide A, Uranga J, Guerrero P, Caba K. Chitosan as a bioactive polymer: Processing, properties and applications. Int J Biol Macromol 2017; 105(2): 1358-68.
[http://dx.doi.org/10.1016/j.ijbiomac.2017.07.087]
[30]
Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev 2016; 99: 28-51.
[http://dx.doi.org/10.1016/j.addr.2015.09.012]
[31]
Gyulai G, Magyar A, Rohonczy J, et al. Preparation and characterization of cationic Pluronic for surface modification and functionalization of polymeric drug delivery nanoparticles. Express Polym Lett 2016; 10(3): 216.
[http://dx.doi.org/10.3144/expresspolymlett.2016.20]
[32]
Badran MM, Mady MM, Ghannam MM, Shakeel F. Preparation and characterization of polymeric nanoparticles surface modified with chitosan for target treatment of colorectal cancer. Int J Biol Macromol 2017; 95: 643-9.
[http://dx.doi.org/10.1016/j.ijbiomac.2016.11.098]
[33]
Abouelmagd SA, Ku YJ, Yeo Y. Low molecular weight chitosan-coated polymeric nanoparticles for sustained and pH-sensitive delivery of paclitaxel. J Drug Target 2015; 23(7-8): 725-35.
[http://dx.doi.org/10.3109/1061186X.2015.1054829]
[34]
Abuchowski A, van Es T, Palczuk NC, Davis FF. Alteration of immunological properties of bovine serum albumin by covalent attachment of polyethylene glycol. J Biol Chem 1977; 252(11): 3578-81.
[35]
Veronese FM, Mero A. The impact of PEGylation on biological therapies. BioDrugs 2008; 22(5): 315-29.
[http://dx.doi.org/10.2165/00063030-200822050-00004]
[36]
Locatelli E, Franchini MC. Biodegradable PLGA-b-PEG polymeric nanoparticles: synthesis, properties, and nanomedical applications as drug delivery system. J Nanopart Res 2012; 14(12): 1316.
[http://dx.doi.org/10.1007/s11051-012-1316-4]
[37]
Zhong Y, Meng F, Deng C, Zhong Z. Ligand-directed active tumor-targeting polymeric nanoparticles for cancer chemotherapy. Biomacromolecules 2014; 15(6): 1955-69.
[http://dx.doi.org/10.1021/bm5003009]
[38]
Jin M, Jin G, Kang L, Chen L, Gao Z, Huang W. Smart polymeric nanoparticles with pH-responsive and PEG-detachable properties for co-delivering paclitaxel and survivin siRNA to enhance antitumor outcomes. Int J Nanomedicine 2018; 13: 2405-26.
[http://dx.doi.org/10.2147/IJN.S161426]
[39]
Sun P, Huang W, Jin M, et al. Chitosan-based nanoparticles for survivin targeted siRNA delivery in breast tumor therapy and preventing its metastasis. Int J Nanomedicine 2016; 11: 4931-45.
[http://dx.doi.org/10.2147/IJN.S105427]
[40]
Patel A, Patel M, Yang X, Mitra AK. Recent advances in protein and peptide drug delivery: a special emphasis on polymeric nanoparticles. Protein Pept Lett 2014; 21(11): 1102-20.
[http://dx.doi.org/10.2174/0929866521666140807114240]
[41]
Lellouche E, Locatelli E, Israel LL, et al. Maghemite-containing PLGA–PEG-based polymeric nanoparticles for siRNA delivery: Toxicity and silencing evaluation. RSC Advances 2017; 7(43): 26912-20.
[http://dx.doi.org/10.1039/C7RA00517B]
[42]
Ho LC, Wu WC, Chang CY, Hsieh HH, Lee CH, Chang HT. Aptamer-conjugated polymeric nanoparticles for the detection of cancer cells through “turn-on” retro-self-quenched fluorescence. Anal Chem 2015; 87(9): 4925-32.
[http://dx.doi.org/10.1021/acs.analchem.5b00569]
[43]
Dhar S, Gu FX, Langer R, Farokhzad OC, Lippard SJ. Targeted delivery of cisplatin to prostate cancer cells by aptamer functionalized Pt (IV) prodrug-PLGA-PEG nanoparticles. Proc Natl Acad Sci USA 2008; 105(45): 17356-61.
[http://dx.doi.org/10.1073/pnas.0809154105]
[44]
Patil YB, Toti US, Khdair A, Ma L, Panyam J. Single-step surface functionalization of polymeric nanoparticles for targeted drug delivery. Biomat 2009; 30(5): 859-66.
[http://dx.doi.org/10.1016/j.biomaterials.2008.09.056]
[45]
Bellocq NC, Pun SH, Jensen GS, Davis ME. Transferrin-containing, cyclodextrin polymer-based particles for tumor-targeted gene delivery. Bioconjug Chem 2003; 14(6): 1122-32.
[http://dx.doi.org/10.1021/bc034125f]
[46]
Mattheolabakis G, Milane L, Singh A, Amiji MM. Hyaluronic acid targeting of CD44 for cancer therapy: From receptor biology to nanomedicine. J Drug Target 2015; 23(7-8): 605-18.
[http://dx.doi.org/10.3109/1061186X.2015.1052072]
[47]
Xiao B, Han MK, Viennois E, et al. Hyaluronic acid-functionalized polymeric nanoparticles for colon cancer-targeted combination chemotherapy. Nanoscale 2015; 7(42): 17745-55.
[http://dx.doi.org/10.1039/C5NR04831A]
[48]
Goodall S, Jones ML, Mahler S. Monoclonal antibody‐targeted polymeric nanoparticles for cancer therapy–future prospects. J Chem Technol Biotechnol 2015; 90(7): 1169-76.
[http://dx.doi.org/10.1002/jctb.4555]
[49]
Luk JM, Wong KF. Monoclonal antibodies as targeting and therapeutic agents: prospects for liver transplantation, hepatitis and hepatocellular carcinoma. Clin Exp Pharmacol Physiol 2006; 33(5-6): 482-8.
[http://dx.doi.org/10.1111/j.1440-1681.2006.04396.x]
[50]
Sperling RA, Parak WJ. Surface modification, functionalization and bioconjugation of colloidal inorganic nanoparticles. Phil Trans R Soc A 1915; 2010(368): 1333-83.
[51]
Gref R, Lück M, Quellec P, et al. ‘Stealth’ corona-core nanoparticles surface modified by polyethylene glycol (PEG): Influences of the corona (PEG chain length and surface density) and of the core composition on phagocytic uptake and plasma protein adsorption. Colloids Surf B Biointerfaces 2000; 18(3-4): 301-13.
[http://dx.doi.org/10.1016/S0927-7765(99)00156-3]
[52]
Hobson B, Denekamp J. Endothelial proliferation in tumours and normal tissues: continuous labelling studies. Br J Cancer 1984; 49(4): 405-13.
[http://dx.doi.org/10.1038/bjc.1984.66]
[53]
Maeda H. The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting. Adv Enzyme Regul 2001; 41: 189-207.
[http://dx.doi.org/10.1016/S0065-2571(00)00013-3]
[54]
Pang L, Pei Y, Uzunalli G, Hyun H, Lyle LT, Yeo Y. Surface modification of polymeric nanoparticles with M2pep peptide for drug delivery to tumor-associated macrophages. Pharm Res 2019; 36(4): 65.
[http://dx.doi.org/10.1007/s11095-019-2596-5]
[55]
Nakanishi T, Fukushima S, Okamoto K, et al. Development of the polymer micelle carrier system for doxorubicin. J Control Release 2001; 74(1-3): 295-302.
[http://dx.doi.org/10.1016/S0168-3659(01)00341-8]
[56]
Joshi G, Kumar A, Sawant K. Bioavailability enhancement, Caco-2 cells uptake and intestinal transport of orally administered lopinavir-loaded PLGA nanoparticles. Drug Deliv 2016; 23(9): 3492-504.
[http://dx.doi.org/10.1080/10717544.2016.1199605]
[57]
Joshi G, Kumar A, Sawant K. Enhanced bioavailability and intestinal uptake of Gemcitabine HCl loaded PLGA nanoparticles after oral delivery. Eur J Pharm Sci 2014; 60: 80-9.
[http://dx.doi.org/10.1016/j.ejps.2014.04.014]
[58]
Pappo J. Generation and characterization of monoclonal antibodies recognizing follicle epithelial M cells in rabbit gut-associated lymphoid tissues. Cell Immunol 1989; 120(1): 31-41.
[http://dx.doi.org/10.1016/0008-8749(89)90172-X]
[59]
Dawson GF, Halbert GW. The in vitro cell association of invasin coated polylactide-co-glycolide nanoparticles. Pharm Res 2000; 17(11): 1420-5.
[http://dx.doi.org/10.1023/A:1007503123620]
[60]
Wang R, Billone PS, Mullett WM. Nanomedicine in action: An overview of cancer nanomedicine on the market and in clinical trials. J Nanomater 2013; 13: 1-12.
[http://dx.doi.org/10.1155/2013/629681]
[61]
Rajagopalan R, Yakhmi JV. Nanotechnological approaches toward cancer chemotherapy Nanostructures for Cancer Therapy. (1st ed): 2017; 211-40.
[62]
Takada N, Kawabe H. Leuprorelin (leuplin, lupron, viadur) in drug discovery in japan Drug Discovery in Japan 2019; 65-84.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy