Biocompatible Polymer Nanoparticles for Drug Delivery Applications in Cancer and Neurodegenerative Disorder Therapies
Abstract
:1. Introduction
2. Nano Biodegradable Polymers
3. Biodegradable NPs in Drug Delivery Systems
3.1. Biodegradable NPs in Cancer Therapy
3.2. Biodegradable NPs Drug Delivery in Neurodegenerative Diseases
4. Conclusions and Outlooks
Funding
Acknowledgments
Conflicts of Interest
References
- Bhatt, P.; Lalani, R.; Mashru, R.; Misra, A. Anti-FSHR antibody Fab’fragment conjugated immunoliposomes loaded with cyclodextrin-paclitaxel complex for improved in vitro efficacy on ovarian cancer cells. Cancer Res. 2016, 76, 2065. [Google Scholar] [CrossRef]
- Vhora, I.; Patil, S.; Bhatt, P.; Misra, A. Protein–and Peptide–Drug Conjugates: An Emerging Drug Delivery Technology. In Advances in Protein Chemistry and Structural Biology; Rossen, D., Ed.; Academic Press-Elsevier: Cambridge, MA, USA, 2015; Volume 98, pp. 1–55. [Google Scholar]
- Sahoo, S.K.; Labhasetwar, V. Nanotech approaches to drug delivery and imaging. Drug Discov. 2003, 8, 1112–1120. [Google Scholar] [CrossRef]
- Duncan, R. The dawning era of polymer therapeutics. Nat. Rev. Drug Discov. 2003, 2, 347–360. [Google Scholar] [CrossRef] [PubMed]
- Moghimi, S.M.; Hunter, A.C.; Murray, J.C. Long-Circulating and Target-Specific Nanoparticles: Theory to Practice. Pharmacol. Rev. 2001, 53, 283–318. [Google Scholar] [PubMed]
- Ulery, B.D.; Nair, L.S.; Laurencin, C.T. Biomedical applications of biodegradable polymers. J. Polym. Sci. B Polym. Phys. 2011, 49, 832–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Makadia, H.K.; Siegel, S.J. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers 2011, 3, 1377–1397. [Google Scholar] [CrossRef]
- Torchilin, V.P. Drug Targeting. Eur. J. Pharm. Sci. 2000, 11, S81–S91. [Google Scholar] [CrossRef]
- Tancini, B.; Tosi, G.; Bortot, B.; Dolcetta, D.; Magini, A.; De Martino, E.; Urbanelli, L.; Ruozi, B.; Forni, F.; Emiliani, C.; et al. Use of polylactide-co-glycolide-nanoparticles for lysosomal delivery of a therapeutic enzyme in glycogenosis type II fibroblasts. J. Nanosci. Nanotechnol. 2015, 15, 2657–2666. [Google Scholar]
- Letchford, K.; Burt, H. A review of the formation and classification of amphiphilic block copolymer nanoparticulate structures: Micelles, nanospheres, nanocapsules and polymersomes. Eur. J. Pharm. Biopharm. 2007, 65, 259–269. [Google Scholar] [CrossRef]
- Ezhilarasi, P.N.; Karthik, P.; Chhanwal, N.; Anandharamakrishnan, C. Nanoencapsulation techniques for food bioactive components: A review. Food Bioprocess Technol. 2013, 6, 628–647. [Google Scholar] [CrossRef]
- Rao, J.P.; Geckeler, K.E. Polymer nanoparticles: Preparation techniques and size-control parameters. Prog. Polym. Sci. 2011, 36, 887–913. [Google Scholar] [CrossRef]
- Joye, I.J.; McClements, D.J. Biopolymer-based nanoparticles and microparticles: Fabrication, characterization, and application. Curr. Opin. Colloid Interface Sci. 2014, 19, 417–427. [Google Scholar] [CrossRef]
- Patel, J.; Amrutiya, J.; Bhatt, P.; Javia, A.; Jain, M.; Misra, A. Targeted delivery of monoclonal antibody conjugated docetaxel loaded PLGA nanoparticles into EGFR overexpressed lung tumour cells. J. Microencapsul. 2018, 35, 204–217. [Google Scholar] [CrossRef] [PubMed]
- Bhatt, P.; Lalani, R.; Vhora, I.; Patil, S.; Amrutiya, J.; Misra, A.; Mashru, R. Liposomes encapsulating native and cyclodextrin enclosed paclitaxel: Enhanced loading efficiency and its pharmacokinetic evaluation. Int. J. Pharm. 2018, 536, 95–107. [Google Scholar] [CrossRef] [PubMed]
- Shoichet, M.S. Polymer scaffolds for biomaterials applications. Macromolecules 2010, 43, 581–591. [Google Scholar] [CrossRef]
- Lu, Y.; Chen, S.C. Micro and nano-fabrication of biodegradable polymers for drug delivery. Adv. Drug Deliv. Rev. 2004, 56, 1621–1633. [Google Scholar] [CrossRef]
- Panyam, J.; Labhasetwar, V. Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Adv. Drug Deliv. Rev. 2003, 55, 329–347. [Google Scholar] [CrossRef]
- Hans, M.L.; Lowman, A.M. Biodegradable nanoparticles for drug delivery and targeting. Curr. Opin. Solid State Mater. Sci. 2002, 6, 319–327. [Google Scholar] [CrossRef]
- Kreuter, J. Drug delivery to the central nervous system by polymeric nanoparticles: What do we know? Adv. Drug Deliv. Rev. 2014, 71, 2–14. [Google Scholar] [CrossRef]
- Ulbrich, K.; Hola, K.; Subr, V.; Bakandritsos, A.; Tucek, J.; Zboril, R. Targeted drug delivery with polymers and magnetic nanoparticles: Covalent and noncovalent approaches, release control, and clinical studies. Chem. Rev. 2016, 116, 5338–5431. [Google Scholar] [CrossRef]
- Parveen, S.; Misra, R.; Sahoo, S.K. Nanoparticles: A boon to drug delivery, therapeutics, diagnostics and imaging. Nanomedicine 2012, 8, 147–166. [Google Scholar] [CrossRef] [PubMed]
- Honey, P.J.; Rijo, J.; Anju, A.; Anoop, K.R. Smart polymers for the controlled delivery of drugs—A concise overview. Acta Pharm. Sin. B 2014, 4, 120–127. [Google Scholar]
- Fleige, E.; Quadir, M.A.; Haag, R. Stimuli-responsive polymeric nanocarriers for the controlled transport of active compounds: Concepts and applications. Adv. Drug Deliv. Rev. 2012, 64, 866–884. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.; Meng, F.; Cheng, R.; Zhong, Z. pH-Sensitive degradable polymersomes for triggered release of anticancer drugs: A comparative study with micelles. J. Control. Release 2010, 142, 40–46. [Google Scholar] [CrossRef] [PubMed]
- Khanal, S.; Adhikari, U.; Rijal, N.P.; Bhattarai, S.R.; Sankar, J.; Bhattarai, N. pH-responsive PLGA nanoparticle for controlled payload delivery of diclofenac sodium. J. Funct. Biomater. 2016, 7, 21. [Google Scholar] [CrossRef] [PubMed]
- Nishiyama, N.; Bae, Y.; Miyata, K.; Fukushima, S.; Kataoka, K. Smart polymeric micelles for gene and drug delivery. Drug Discov. Today 2005, 2, 21–26. [Google Scholar] [CrossRef] [PubMed]
- Sawant, R.M.; Hurley, J.P.; Salmaso, S.; Kale, A.; Tolcheva, E.; Levchenko, T.S.; Torchilin, V.P. “SMART” Drug Delivery Systems: Double-Targeted pH-Responsive Pharmaceutical Nanocarriers. Bioconjugate Chem. 2006, 17, 943–949. [Google Scholar] [CrossRef] [PubMed]
- Torchilin, V.P. Structure and design of polymeric surfactant-based drug delivery systems. J. Control. Release 2001, 73, 137–172. [Google Scholar] [CrossRef]
- Emerich, D.F.; Thanos, C.G. The pinpoint promise of nanoparticle-based drug delivery and molecular diagnosis. Biomol. Eng. 2006, 23, 171–184. [Google Scholar] [CrossRef]
- Farokhzad, O.C.; Langer, R. Nanomedicine: Developing smarter therapeutic and diagnostic modalities. Adv. Drug Deliv. Rev. 2006, 58, 1456–1459. [Google Scholar] [CrossRef]
- Beck, R.C.R.; Chaves, P.S.; Goyanes, A.; Vukosavljevic, B.; Buanz, A.; Windbergs, M.; Basit, A.W.; Gaisford, S. 3D printed tablets loaded with polymeric nanocapsules: An innovative approach to produce customized drug delivery systems. Int. J. Pharm. 2017, 528, 268–279. [Google Scholar] [CrossRef]
- Gaumet, M.; Vargas, A.; Gurny, R.; Delie, F. Nanoparticles for drug delivery: The need for precision in reporting particle size parameters. Eur. J. Pharm. Biopharm. 2008, 69, 1–9. [Google Scholar] [CrossRef]
- Wissing, S.A.; Kayser, O.; Müller, R.H. Solid lipid nanoparticles for parenteral drug delivery. Adv. Drug Deliv. Rev. 2004, 56, 1257–1272. [Google Scholar] [CrossRef] [PubMed]
- Duncan, R. Polymer conjugates as anticancer nanomedicines. Nat. Rev. Cancer 2006, 6, 688–701. [Google Scholar] [CrossRef] [PubMed]
- Lammers, T.; Subr, V.; Ulbrich, K.; Hennink, W.E.; Storm, G.; Kiessling, F. Polymeric nanomedicines for image-guided drug delivery and tumor-targeted combination therapy. Nano Today 2010, 5, 197–212. [Google Scholar] [CrossRef]
- Grund, S.; Bauer, M.; Fischer, D. Polymers in drug delivery—State of the art and future trends. Adv. Eng. Mater. 2011, 13, B61–B87. [Google Scholar] [CrossRef]
- Gref, R.; Lück, M.; Quellec, P.; Marchand, M.; Dellacherie, E.; Harnisch, S.; Blunk, T.; Müller, R.H. ‘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 2000, 18, 301–313. [Google Scholar] [CrossRef]
- Park, J.; Fong, P.M.; Lu, J.; Russell, K.S.; Booth, C.J.; Saltzman, W.M.; Fahmy, T.M. PEGylated PLGA nanoparticles for the improved delivery of doxorubicin. Nanomedicine 2009, 5, 410–418. [Google Scholar] [CrossRef] [Green Version]
- Krishna, R.; Mayer, L.D. Multidrug resistance (MDR) in cancer: Mechanisms, reversal using modulators of MDR and the role of MDR modulators in influencing the pharmacokinetics of anticancer drugs. Eur. J. Pharm. Sci. 2000, 11, 265–283. [Google Scholar] [CrossRef]
- Çırpanlı, Y.; Allard, E.; Passirani, C.; Bilensoy, E.; Lemaire, L.; Çalış, S.; Benoit, J.P. Antitumoral activity of camptothecin-loaded nanoparticles in 9L rat glioma model. Int. J. Pharm. 2011, 403, 201–206. [Google Scholar] [CrossRef]
- Yuan, J.D.; ZhuGe, D.L.; Tong, M.Q.; Lin, M.T.; Xu, X.F.; Tang, X.; Zhao, Y.Z.; Xu, H.L. pH-sensitive polymeric nanoparticles of mPEG-PLGA-PGlu with hybrid core for simultaneous encapsulation of curcumin and doxorubicin to kill the heterogeneous tumour cells in breast cancer. Artif. Cells Nanomed. Biotechnol. 2018, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Hu, D.; Chen, L.; Qu, Y.; Peng, J.; Chu, B.; Shi, K.; Hao, Y.; Zhong, L.; Wang, M.; Qian, Z. Oxygen-generating hybrid polymeric nanoparticles with encapsulated doxorubicin and chlorin e6 for trimodal imaging-guided combined chemo-photodynamic therapy. Theranostics 2018, 8, 1558–1574. [Google Scholar] [CrossRef] [PubMed]
- Ahmad, N.; Ahmad, R.; Alam, M.A.; Ahmad, F.J. Enhancement of oral bioavailability of doxorubicin through surface modified biodegradable polymeric nanoparticles. Chem. Cent. J. 2018, 12, 65. [Google Scholar] [CrossRef] [PubMed]
- Malinovskaya, Y.; Melnikov, P.; Baklaushev, V.; Gabashvili, A.; Osipova, N.; Mantrov, S.; Ermolenko, Y.; Maksimenko, O.; Gorshkola, M.; Balabanyan, V.; et al. Delivery of doxorubicin-loaded PLGA nanoparticles into U87 human glioblastoma cells. Int. J. Pharm. 2017, 524, 77–90. [Google Scholar] [CrossRef] [PubMed]
- Soma, C.E.; Dubernet, C.; Bentolila, D.; Benita, S.; Couvreur, P. Reversion of multidrug resistance by co-encapsulation of doxorubicin and cyclosporin A in polyalkylcyanoacrylate nanoparticles. Biomaterials 2002, 21, 1–7. [Google Scholar] [CrossRef]
- Hekmatara, T.; Bernreuther, C.; Khalansky, A.S.; Theisen, A.; Weissenberger, J.; Matschke, J.; Gelperina, S.; Kreuter, J.; Glatzel, M. Efficient systemic therapy of rat glioblastoma by nanoparticle-bound doxorubicin is due to antiangiogenic effects. Clin. Neuropathol. 2009, 28, 153–164. [Google Scholar] [CrossRef] [PubMed]
- Hu, J.; Fu, S.; Peng, Q.; Han, Y.; Xie, J.; Zan, N.; Chen, Y.; Fan, J. Paclitaxel-loaded polymeric nanoparticles combined with chronomodulated chemotherapy on lung cancer: In vitro and in vivo evaluation. Int. J. Pharm. 2017, 516, 313–322. [Google Scholar] [CrossRef] [PubMed]
- 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. Nanomed. 2018, 13, 2405–2423. [Google Scholar] [CrossRef]
- Guo, J.; Gao, X.; Su, L.; Xia, H.; Gu, G.; Pang, Z.; Jiang, X.; Yao, L.; Chen, J.; Chen, H. Aptamer-functionalized PEG–PLGA nanoparticles for enhanced anti-glioma drug delivery. Biomaterials 2011, 32, 8010–8020. [Google Scholar] [CrossRef]
- Shafiei-Irannejad, V.; Samadi, N.; Salehi, R.; Yousefi, B.; Rahimi, M.; Akbarzadeh, A.; Zarghami, N. Reversion of Multidrug Resistance by Co-Encapsulation of Doxorubicin and Metformin in Poly (lactide-co-glycolide)-d-α-tocopheryl Polyethylene Glycol 1000 Succinate Nanoparticles. Pharm. Res. 2018, 35, 119. [Google Scholar] [CrossRef]
- Bae, Y.H. Drug Targeting and Tumor Heterogeneity. J. Control. Release 2009, 133, 2–3. [Google Scholar] [CrossRef] [PubMed]
- Danhier, F.; Feron, O.; Préat, V. To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J. Control. Release 2010, 148, 135–146. [Google Scholar] [CrossRef] [PubMed]
- Torchilin, V. Tumor delivery of macromolecular drugs based on the EPR effect. Adv. Drug Deliv. Rev. 2011, 63, 131–135. [Google Scholar] [CrossRef] [PubMed]
- Adjei, I.M.; Blanka, S. Modulation of the tumor microenvironment for cancer treatment: A biomaterials approach. J. Funct. Biomater. 2015, 6, 81–103. [Google Scholar] [CrossRef] [PubMed]
- Adams, G.P.; Schier, R.; McCall, A.M.; Simmons, H.H.; Horak, E.M.; Alpaugh, R.K.; Marks, J.D.; Weiner, L.M. High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules. Cancer Res. 2001, 61, 4750–4755. [Google Scholar] [PubMed]
- Sanna, V.; Singh, C.K.; Jashari, R.; Adhami, V.M.; Chamcheu, J.C.; Rady, I.; Sechi, M.; Mukhtar, H.; Siddiqui, I.A. Targeted nanoparticles encapsulating (−)-epigallocatechin-3-gallate for prostate cancer prevention and therapy. Sci. Rep. 2017, 7, 41573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kong, N.; Deng, M.; Sun, X.N.; Chen, Y.D.; Sui, X.B. Polydopamine-Functionalized CA-(PCL-ran-PLA) Nanoparticles for Target Delivery of Docetaxel and Chemo-photothermal Therapy of Breast Cancer. Front. Pharmacol. 2018, 9, 125. [Google Scholar] [CrossRef]
- Fan, Y.; Moon, J.J. Nanoparticle drug delivery systems designed to improve cancer vaccines and immunotherapy. Vaccines 2015, 3, 662–685. [Google Scholar] [CrossRef]
- Kim, H.; Niu, L.; Larson, P.; Kucaba, T.A.; Murphy, K.A.; James, B.R.; Ferguson, D.M.; Griffith, T.S.; Panyam, J. Polymeric nanoparticles encapsulating novel TLR7/8 agonists as immunostimulatory adjuvants for enhanced cancer immunotherapy. Biomaterials 2018, 164, 38–53. [Google Scholar] [CrossRef]
- Roy, A.; Singh, M.S.; Upadhyay, P.; Bhaskar, S. Combined chemo-immunotherapy as a prospective strategy to combat cancer: A nanoparticle based approach. Mol. Pharm. 2010, 7, 1778–1788. [Google Scholar] [CrossRef]
- Polchi, A.; Magini, A.; Mazuryk, J.; Tancini, B.; Gapiński, J.; Patkowski, A.; Giovagnoli, S.; Emiliani, C. Rapamycin loaded solid lipid nanoparticles as a new tool to deliver mTOR inhibitors: Formulation and in vitro characterization. Nanomaterials 2016, 6, 87. [Google Scholar] [CrossRef] [PubMed]
- Cacciatore, I.; Ciulla, M.; Fornasari, E.; Marinelli, L.; Di Stefano, A. Solid lipid nanoparticles as a drug delivery system for the treatment of neurodegenerative diseases. Expert Opin. Drug Deliv. 2016, 13, 1121–1131. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Li, H.; Luo, G.; Liu, Q.; Wang, Y. Pharmacokinetics and biodistribution of surface modification polymeric nanoparticles. Arch. Pharm. Res. 2008, 31, 547–554. [Google Scholar] [CrossRef] [PubMed]
- Rao, K.S.; Reddy, M.K.; Horning, J.L.; Labhasetwar, V. TAT-conjugated nanoparticles for the CNS delivery of anti-HIV drugs. Biomaterials 2008, 29, 4429–4438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kulkarni, S.A.; Feng, S.S. Effects of surface modification on delivery efficiency of biodegradable nanoparticles across the blood–brain barrier. Nanomedicine 2011, 6, 377–394. [Google Scholar] [CrossRef] [PubMed]
- Geldenhuys, W.; Mbimba, T.; Bui, T.; Harrison, K.; Sutariya, V. Brain-targeted delivery of paclitaxel using glutathione-coated nanoparticles for brain cancers. J. Drug Target 2011, 19, 837–845. [Google Scholar] [CrossRef] [PubMed]
- Tosi, G.; Costantino, L.; Rivasi, F.; Ruozi, B.; Leo, E.; Vergoni, A.V.; Tacchi, R.; Bertolini, A.; Vandelli, M.A.; Forni, F. Targeting the central nervous system: In vivo experiments with peptide-derivatized nanoparticles loaded with Loperamide and Rhodamine-123. J. Control. Release 2007, 122, 1–9. [Google Scholar] [CrossRef]
- Vergoni, A.V.; Tosi, G.; Tacchi, R.; Vandelli, M.A.; Bertolini, A.; Costantino, L. Nanoparticles as drug delivery agents specific for CNS: In vivo biodistribution. Nanomedicine 2009, 5, 369–377. [Google Scholar] [CrossRef]
- Zhang, C.; Wan, X.; Zheng, X.; Shao, X.; Liu, Q.; Zhang, Q.; Qian, Y. Dual-functional nanoparticles targeting amyloid plaques in the brains of Alzheimer’s disease mice. Biomaterials 2014, 35, 456–465. [Google Scholar] [CrossRef]
- Sánchez-López, E.; Ettcheto, M.; Egea, M.A.; Espina, M.; Cano, A.; Calpena, A.C.; Camins, A.; Carmona, N.; Silva, A.M.; Souto, E.B.; et al. Memantine loaded PLGA PEGylated nanoparticles for Alzheimer’s disease: In vitro and in vivo characterization. J. Nanobiotechnol. 2018, 16, 32. [Google Scholar] [CrossRef]
- Barcia, E.; Boeva, L.; García-García, L.; Slowing, K.; Fernández-Carballido, A.; Casanova, Y.; Negro, S. Nanotechnology-based drug delivery of ropinirole for Parkinson’s disease. Drug Deliv. 2017, 24, 1112–1123. [Google Scholar] [CrossRef] [PubMed]
- Bourdenx, M.; Daniel, J.; Genin, E.; Soria, F.N.; Blanchard-Desce, M.; Bezard, E.; Dehay, B. Nanoparticles restore lysosomal acidification defects: Implications for Parkinson and other lysosomal-related diseases. Autophagy 2016, 12, 472–483. [Google Scholar] [CrossRef]
- Barbara, R.; Belletti, D.; Pederzoli, F.; Masoni, M.; Keller, J.; Ballestrazzi, A.; Vandelli, M.A.; Tosi, G.; Grabrucker, A.M. Novel Curcumin loaded nanoparticles engineered for Blood-Brain Barrier crossing and able to disrupt Abeta aggregates. Int. J. Pharm. 2017, 526, 413–424. [Google Scholar] [CrossRef]
- Huang, N.; Lu, S.; Liu, X.G.; Zhu, J.; Wang, Y.J.; Liu, R.T. PLGA nanoparticles modified with a BBB-penetrating peptide co-delivering Aβ generation inhibitor and curcumin attenuate memory deficits and neuropathology in Alzheimer’s disease mice. Oncotarget 2017, 8, 81001–81013. [Google Scholar] [PubMed]
- Sun, D.; Li, N.; Zhang, W.; Zhao, Z.; Mou, Z.; Huang, D.; Liu, J.; Wang, W. Design of PLGA-functionalized quercetin nanoparticles for potential use in Alzheimer’s disease. Colloids Surf. B Biointerfaces 2016, 148, 116–129. [Google Scholar] [CrossRef]
- Kuo, Y.C.; Tsai, H.C. Rosmarinic acid- and curcumin-loaded polyacrylamide-cardiolipin-poly(lactide-co-glycolide) nanoparticles with conjugated 83-14 monoclonal antibody to protect β-amyloid-insulted neurons. Mater. Sci. Eng. C Mater. Biol. Appl. 2018, 91, 445–457. [Google Scholar] [CrossRef]
- Kuo, Y.C.; Rajesh, R. Targeted delivery of rosmarinic acid across the blood-brain barrier for neuronal rescue using polyacrylamide-chitosan-poly(lactide-co-glycolide) nanoparticles with surface cross-reacting material 197 and apolipoprotein E. Int. J. Pharm. 2017, 528, 228–241. [Google Scholar] [CrossRef] [PubMed]
- Martin-Banderas, L.; Holgado, M.A.; Durán-Lobato, M.; Infante, J.J.; Fernández-Arévalo, M. Role of nanotechnology for enzyme replacement therapy in lysosomal diseases. A focus on Gaucher’s disease. Curr. Med. Chem. 2016, 23, 929–952. [Google Scholar] [CrossRef]
- Urbanelli, L.; Magini, A.; Polchi, A.; Polidoro, M.; Emiliani, C. Recent developments in therapeutic approaches for lysosomal storage diseases. Recent Pat. CNS Drug Discov. 2011, 6, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Martino, S.; Emiliani, C.; Tancini, B.; Severini, G.M.; Chigorno, V.; Bordignon, C.; Sonnino, S.; Orlacchio, A. Absence of metabolic cross-correction in Tay-Sachs cells: Implications for gene therapy. J. Biolol. Chem. 2002, 277, 20177–20184. [Google Scholar] [CrossRef]
- Martino, S.; Marconi, P.; Tancini, B.; Dolcetta, D.; De Angelis, M.C.; Montanucci, P.; Bregola, G.; Sandhoff, K.; Bordignon, C.; Emiliani, C.; et al. A direct gene transfer strategy via brain internal capsule reverses the biochemical defect in Tay–Sachs disease. Hum. Mol. Genet. 2005, 14, 2113–2123. [Google Scholar] [CrossRef] [PubMed]
- Arfi, A.; Bourgoin, C.; Basso, L.; Emiliani, C.; Tancini, B.; Chigorno, V.; Li, Y.T.; Orlacchio, A.; Poenaru, L.; Sonnino, S.; et al. Bicistronic lentiviral vector corrects β-hexosaminidase deficiency in transduced and cross-corrected human Sandhoff fibroblasts. Neurobiol. Dis. 2005, 20, 583–593. [Google Scholar] [CrossRef] [PubMed]
- Bourgoin, C.; Emiliani, C.; Kremer, E.J.; Gelot, A.; Tancini, B.; Gravel, R.A.; Drugan, C.; Orlacchio, A.; Poenaru, L.; Caillaud, C. Widespread distribution of β-hexosaminidase activity in the brain of a Sandhoff mouse model after coinjection of adenoviral vector and mannitol. Gene Ther. 2003, 10, 1841–1849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, P.; Li, Y.; Nikolaishvili-Feinberg, N.; Scesa, G.; Bi, Y.; Pan, D.; Moore, D.; Bongarzone, E.R.; Sands, M.S.; Miller, R.; et al. Hematopoietic Stem cell transplantation and lentiviral vector-based gene therapy for Krabbe’s disease: Present convictions and future prospects. J. Neurosci. Res. 2016, 94, 1152–1168. [Google Scholar] [CrossRef] [PubMed]
- Biffi, A. Hematopoietic stem cell gene therapy for storage disease: Current and new indications. Mol. Ther. 2017, 25, 1155–1162. [Google Scholar] [CrossRef]
- Ohashi, T. Enzyme replacement therapy for lysosomal storage diseases. Pediatr. Endocrinol. Rev. (PER) 2012, 10, 26–34. [Google Scholar]
- Muro, S. Strategies for delivery of therapeutics into the central nervous system for treatment of lysosomal storage disorders. Drug. Deliv. Transl. Res. 2012, 2, 169–186. [Google Scholar] [CrossRef] [Green Version]
- Muro, S. New biotechnological and nanomedicine strategies for treatment of lysosomal storage disorders. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2010, 2, 189–204. [Google Scholar] [CrossRef] [Green Version]
- Donida, B.; Tauffner, B.; Raabe, M.; Immich, M.F.; de Farias, M.A.; de Sá Coutinho, D.; Zaffari Machado, A.; Kessler, R.G.; Portugal, R.V.; Bernardi, A.; et al. Monoolein-based nanoparticles for drug delivery to the central nervous system: A platform for lysosomal storage disorder treatment. Eur. J. Pharm. Biopharm. 2018, 133, 96–103. [Google Scholar] [CrossRef]
- Salvalaio, M.; Rigon, L.; Belletti, D.; D’Avanzo, F.; Pederzoli, F.; Ruozi, B.; Marin, O.; Vandelli, M.A.; Forni, F.; Scarpa, M.; et al. Targeted polymeric nanoparticles for brain delivery of high molecular weight molecules in lysosomal storage disorders. PLoS ONE 2016, 11, e0156452. [Google Scholar] [CrossRef]
- Schuster, T.; Mühlstein, A.; Yaghootfam, C.; Maksimenko, O.; Shipulo, E.; Gelperina, S.; Kreuter, J.; Gieselmann, V.; Matzner, U. Potential of surfactant-coated nanoparticles to improve brain delivery of arylsulfatase A. J. Control. Release 2017, 253, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Woodrow, K.A.; Cu, Y.; Booth, C.J.; Saucier-Sawyer, J.K.; Wood, M.J.; Saltzman, W.M. Intravaginal gene silencing using biodegradable polymer nanoparticles densely loaded with small-interfering RNA. Nat. Mater. 2009, 8, 526–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Demento, S.L.; Eisenbart, S.C.; Foellmer, H.G.; Platt, C.; Caplan, M.J.; Saltzman, W.M.; Mellman, I.; Ledizet, M.; Fikrig, E.; Flavell, R.A.; et al. Inflammasome-activating nanoparticles as modular systems for optimizing vaccine efficacy. Vaccine 2009, 27, 3013–3021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fahmy, T.M.; Fong, P.M.; Park, J.; Constable, T.; Saltzman, W.M. Nanosystems for simultaneous imaging and drug delivery to T cells. AAPS J. 2007, 9, E171–E179. [Google Scholar] [CrossRef] [PubMed]
- Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; del Pilar Rodriguez-Torres, M.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef] [PubMed]
- Skotland, T.; Iversen, T.G.; Sandvig, K. Development of nanoparticles for clinical use. Nanomedicine 2014, 9, 1295–1299. [Google Scholar] [CrossRef] [PubMed]
- Gendelman, H.E.; Anantharam, V.; Bronich, T.; Ghaisas, S.; Jin, H.; Kanthasamy, A.G.; Liu, X.; McMillan, J.E.; Monsley, R.L.; Narasimhan, B.; Mallapragada, S.K. Nanoneuromedicines for degenerative, inflammatory, and infectious nervous system diseases. Nanomedicine 2015, 11, 751–767. [Google Scholar] [CrossRef] [Green Version]
Drug | Polymer | Loading Mode | Reference |
---|---|---|---|
CPT | PCL-PEG | Entrapment | [41] |
DOX-CUR | mPEG-PLGA-PGlu | Encapsulation | [42] |
DOX-Chlorin e6-MnO2 | PCLLA-PEG-PCLLA | Encapsulation | [43] |
DOX | PLGA | Entrapment | [44] |
DOX | PLGA-Cyanine5.5 | Encapsulation | [45]] |
DOX-Metformin | PLGA-TPGS | Encapsulation | [46] |
DOX | PBCA | Encapsulation | [47] |
PTX | PCL-PEG-PCL | Encapsulation | [48] |
PTX | PEI-PLA | Entrapment | [49] |
PTX | PLGA-PEG | Encapsulation | [50] |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Calzoni, E.; Cesaretti, A.; Polchi, A.; Di Michele, A.; Tancini, B.; Emiliani, C. Biocompatible Polymer Nanoparticles for Drug Delivery Applications in Cancer and Neurodegenerative Disorder Therapies. J. Funct. Biomater. 2019, 10, 4. https://doi.org/10.3390/jfb10010004
Calzoni E, Cesaretti A, Polchi A, Di Michele A, Tancini B, Emiliani C. Biocompatible Polymer Nanoparticles for Drug Delivery Applications in Cancer and Neurodegenerative Disorder Therapies. Journal of Functional Biomaterials. 2019; 10(1):4. https://doi.org/10.3390/jfb10010004
Chicago/Turabian StyleCalzoni, Eleonora, Alessio Cesaretti, Alice Polchi, Alessandro Di Michele, Brunella Tancini, and Carla Emiliani. 2019. "Biocompatible Polymer Nanoparticles for Drug Delivery Applications in Cancer and Neurodegenerative Disorder Therapies" Journal of Functional Biomaterials 10, no. 1: 4. https://doi.org/10.3390/jfb10010004
APA StyleCalzoni, E., Cesaretti, A., Polchi, A., Di Michele, A., Tancini, B., & Emiliani, C. (2019). Biocompatible Polymer Nanoparticles for Drug Delivery Applications in Cancer and Neurodegenerative Disorder Therapies. Journal of Functional Biomaterials, 10(1), 4. https://doi.org/10.3390/jfb10010004