Since they were first proposed as nonviral transfection agents for their gene-carrying capacity, magnetic nanoparticles have been studied thoroughly, both in vitro and in vivo. Great effort has been made to manufacture biocompatible magnetic nanoparticles for use in the theragnosis of cancer and other diseases. Here we survey recent advances in the study of magnetic nanoparticles, as well as the polymers and other coating layers currently available for gene therapy, their synthesis, and bioconjugation processes. In addition, we review several gene therapy models based on magnetic nanoparticles. 1. Introduction 1.1. Cancer & Current Therapy Although huge efforts have led to advances in cancer treatment, this multifactorial and heterogeneous disease is still one of the major causes of death in the majority of countries [1–4]. Several factors influence the high death rate of cancer patients around the world, such as the genetic and phenotypic heterogeneity and the highly unstable genome of cancer cells, which ultimately leads to continuously emerging, nonheterogeneous cells from the tumor nest, and subsequent metastases [5]. Current cancer therapy encompasses a wide variety of treatments from systemic cytostatics to targeted therapy agents, most still in development phases, such as kinase inhibitors [6–9], antibodies [10, 11], small molecules, or cell- and antigen-based immunotherapies [12, 13]. Nevertheless, most approved therapeutics require systemic administration, which increases their toxicity and other clinical complications. One factor that eventually contributes to therapeutic toxicity is nonspecificity. Guidance of drugs to the desired tissue and specific target recognition are therefore major concerns faced by cancer researchers seeking less toxic side effects and improved efficiency of therapy. Targeted strategies include ligand-receptor binding, antibody-antigen specificity, and some other forms of active targeting. For instance, several antibodies have been developed that are directed to specific tumor-associated antigens, either expressed uniquely by tumor cells or overexpressed compared with healthy tissue; this is the case of the HER2-specific antibody Herceptin [10], the VEGF-specific antibody Bevacizumab [11], and ganglioside-specific antibodies such as 14F7 [14]. Nanotechnology, and in particular a magnetic nanoparticle-based approach, is a promising tool for the guidance of therapeutic agents into tumor tissues. 1.2. Nanotechnology in Cancer Theragnosis Use of nanotechnology is widespread in several fields of biomedicine due to its
References
[1]
K. A. Lowe, V. M. Chia, A. Taylor et al., “An international assessment of ovarian cancer incidence and mortality,” Gynecologic Oncology, vol. 130, no. 1, pp. 107–114, 2013.
[2]
D. Schottenfeld, J. L. Beebe-Dimmer, P. A. Buffler, and G. S. Omenn, “Current perspective on the global and United States cancer burden attributable to lifestyle and environmental risk factors,” Annual Review of Public Health, vol. 34, pp. 97–117, 2013.
[3]
J. Amaro, M. Severo, S. Vilela et al., “Patterns of breast cancer mortality trends in Europe,” The Breast, vol. 22, no. 3, pp. 244–253, 2013.
[4]
S. Walters, C. Maringe, J. Butler et al., “Breast cancer survival and stage at diagnosis in Australia, Canada, Denmark, Norway, Sweden and the UK, 2000–2007: a population-based study,” British Journal of Cancer, vol. 108, no. 5, pp. 1195–1208, 2013.
[5]
J. J. Salk, E. J. Fox, and L. A. Loeb, “Mutational heterogeneity in human cancers: origin and consequences,” Annual Review of Pathology, vol. 5, pp. 51–75, 2010.
[6]
S. Vinayak and R. W. Carlson, “mTOR inhibitors in the treatment of breast cancer,” Oncology, vol. 27, no. 1, pp. 38–44, 2013.
[7]
A. L. Ho, R. K. Grewal, R. Leboeuf et al., “Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer,” The New England Journal of Medicine, vol. 368, no. 7, pp. 623–632, 2013.
[8]
E. I. Heath, J. Infante, L. D. Lewis et al., “A randomized, double-blind, placebo-controlled study to evaluate the effect of repeated oral doses of pazopanib on cardiac conduction in patients with solid tumors,” Cancer Chemotherapy and Pharmacology, vol. 71, no. 3, pp. 565–573, 2013.
[9]
J. W. Welsh, R. Komaki, A. Amini et al., “Phase II trial of erlotinib plus concurrent whole-brain radiation therapy for patients with brain metastases from non-small-cell lung cancer,” Journal of Clinical Oncology, vol. 31, no. 7, pp. 895–902, 2013.
[10]
G. Ismael, R. Hegg, S. Muehlbauer et al., “Subcutaneous versus intravenous administration of (neo)adjuvant trastuzumab in patients with HER2-positive, clinical stage I-III breast cancer (HannaH study): a phase 3, open-label, multicentre, randomised trial,” The Lancet Oncology, vol. 13, no. 9, pp. 869–878, 2012.
[11]
R. Tielen, C. Verhoef, F. van Coevorden et al., “Surgical treatment of locally advanced, non-metastatic, gastrointestinal stromal tumours after treatment with imatinib,” European Journal of Surgical Oncology, vol. 39, no. 2, pp. 150–155, 2013.
[12]
W. Li, L. P. Xu, D. I. Zhao L et al., “Cytokine-induced killer cell therapy for advanced pancreatic adenocarcinoma: a case report and review of the literature,” Oncology Letters, vol. 5, no. 4, pp. 1427–1429, 2013.
[13]
V. Mulens, A. De La Torre, P. Marinello et al., “Immunogenicity and safety of a NeuGcGM3 based cancer vaccine: results from a controlled study in metastatic breast cancer patients,” Human Vaccines, vol. 6, no. 9, pp. 736–744, 2010.
[14]
Y. Fernández-Marrero, L. Roque-Navarro, T. Hernández et al., “A cytotoxic humanized anti-ganglioside antibody produced in a murine cell line defective of N-glycolylated-glycoconjugates,” Immunobiology, vol. 216, no. 12, pp. 1239–1247, 2011.
[15]
J. Chomoucka, J. Drbohlavova, D. Huska, V. Adam, R. Kizek, and J. Hubalek, “Magnetic nanoparticles and targeted drug delivering,” Pharmacological Research, vol. 62, no. 2, pp. 144–149, 2010.
[16]
J. Klostergaard and C. E. Seeney, “Magnetic nanovectors for drug delivery,” Nanomedicine, vol. 8, pp. S37–S50, 2012.
[17]
T. D. Schladt, K. Schneider, H. Schild, and W. Tremel, “Synthesis and bio-functionalization of magnetic nanoparticles for medical diagnosis and treatment,” Dalton Transactions, vol. 40, no. 24, pp. 6315–6343, 2011.
[18]
M. J. Ruedas-Rama, J. D. Walters, A. Orte, and E. A. H. Hall, “Fluorescent nanoparticles for intracellular sensing: a review,” Analytica Chimica Acta, vol. 751, pp. 1–23, 2012.
[19]
Wahajuddin and S. Arora, “Superparamagnetic iron oxide nanoparticles: magnetic nanoplatforms as drug carriers,” International Journal of Nanomedicine, vol. 7, pp. 3445–3471, 2012.
[20]
J. M. Vargas, E. Lima Jr., R. D. Zysler, J. G. S. Duque, E. De Biasi, and M. Knobel, “Effective anisotropy field variation of magnetite nanoparticles with size reduction,” European Physical Journal B, vol. 64, no. 2, pp. 211–218, 2008.
[21]
H. Koo, M. S. Huh, I.-C. Sun et al., “In vivo targeted delivery of nanoparticles for theranosis,” Accounts of Chemical Research, vol. 44, no. 10, pp. 1018–1028, 2011.
[22]
T. E. Mccann, N. Kosaka, B. Turkbey, M. Mitsunaga, P. L. Choyke, and H. Kobayashi, “Molecular imaging of tumor invasion and metastases: the role of MRI,” NMR in Biomedicine, vol. 24, no. 6, pp. 561–568, 2011.
[23]
D. A. Green, M. Durand, N. Gumpeni et al., “Role of magnetic resonance imaging in bladder cancer: current status and emerging techniques,” BJU International, vol. 110, no. 10, pp. 1463–1470, 2012.
[24]
M. Sarparanta, L. M. Bimbo, J. Rytko?nen et al., “Intravenous delivery of hydrophobin-functionalized porous silicon nanoparticles: stability, plasma protein adsorption and biodistribution,” Molecular Pharmaceutics, vol. 9, no. 3, pp. 654–663, 2012.
[25]
Y. Zhao, C. Wang, L. Wang et al., “A frustrating problem: accelerated blood clearance of PEGylated solid lipid nanoparticles following subcutaneous injection in rat,” European Journal of Pharmaceutics and Biopharmaceutics, vol. 81, no. 3, pp. 506–513, 2012.
[26]
M. Baek, H. E. Chung, J. Yu et al., “Pharmacokinetics, tissue distribution, and excretion of zinc oxide nanoparticles,” International Journal of Nanomedicine, vol. 7, pp. 3081–3097, 2012.
[27]
J. E. Zuckerman, C. H. J. Choi, H. Han, and M. E. Davis, “Polycation-siRNA nanoparticles can disassemble at the kidney glomerular basement membrane,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 8, pp. 3137–3142, 2012.
[28]
B. Naeye, H. Deschout, V. Caveliers et al., “In vivo disassembly of IV administered siRNA matrix nanoparticles at the renal filtration barrier,” Biomaterials, vol. 34, no. 9, pp. 2350–2358, 2013.
[29]
H. S. Choi, W. Liu, P. Misra et al., “Renal clearance of quantum dots,” Nature Biotechnology, vol. 25, no. 10, pp. 1165–1170, 2007.
[30]
D. Venturoli and B. Rippe, “Ficoll and dextran vs. globular proteins as probes for testing glomerular permselectivity: effects of molecular size, shape, charge, and deformability,” American Journal of Physiology: Renal Physiology, vol. 288, no. 4, pp. F605–F613, 2005.
[31]
A. T. Florence and N. Hussain, “Transcytosis of nanoparticle and dendrimer delivery systems: evolving vistas,” Advanced Drug Delivery Reviews, vol. 50, supplement 1, pp. S69–S89, 2001.
[32]
B. He, Z. Jia, W. Du et al., “The transport pathways of polymer nanoparticles in MDCK epithelial cells,” Biomaterials, vol. 34, no. 17, pp. 4309–4326, 2013.
[33]
H. Hillaireau and P. Couvreur, “Nanocarriers' entry into the cell: relevance to drug delivery,” Cellular and Molecular Life Sciences, vol. 66, no. 17, pp. 2873–2896, 2009.
[34]
E. Fr?hlich, “The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles,” International Journal of Nanomedicine, vol. 7, pp. 5577–5591, 2012.
[35]
A. Lesniak, A. Salvati, M. J. Santos-Martinez, M. W. Radomski, K. A. Dawson, and C. ?berg, “Nanoparticle adhesion to the cell membrane and its effect on nanoparticle uptake efficiency,” Journal of the American Chemical Society, vol. 135, no. 4, pp. 1438–1444, 2013.
[36]
Z. J. Deng, M. Liang, I. Toth, M. Monteiro, and R. F. Minchin, “Plasma protein binding of positively and negatively charged polymer-coated gold nanoparticles elicits different biological responses,” Nanotoxicology, vol. 7, pp. 314–322, 2013.
[37]
A. Albanese, P. S. Tang, and W. C. W. Chan, “The effect of nanoparticle size, shape, and surface chemistry on biological systems,” Annual Review of Biomedical Engineering, vol. 14, pp. 1–16, 2012.
[38]
H. Maeda, “Vascular permeability in cancer and infection as related to macromolecular drug delivery, with emphasis on the EPR effect for tumor-selective drug targeting,” Proceedings of the Japan Academy B, vol. 88, no. 3, pp. 53–71, 2012.
[39]
N. T. Kimura, S. Taniguchi, K. Aoki, and T. Baba, “Selective localization and growth of Bifidobacterium bifidum in mouse tumors following intravenous administration,” Cancer Research, vol. 40, no. 6, pp. 2061–2068, 1980.
[40]
A. J. Giustini, A. A. Petryk, and P. J. Hoopes, “Ionizing radiation increases systemic nanoparticle tumor accumulation,” Nanomedicine, vol. 8, no. 6, pp. 818–821, 2012.
[41]
H. Gao, J. Xiong, T. Cheng et al., “In vivo biodistribution of mixed shell micelles with tunable hydrophilic/hydrophobic surface,” Biomacromolecules, vol. 14, no. 2, pp. 460–467, 2013.
[42]
V. K. Lamer and R. H. Dinegar, “Theory, production and mechanism of formation of monodispersed hydrosols,” Journal of the American Chemical Society, vol. 72, no. 11, pp. 4847–4854, 1950.
[43]
T. Sugimoto and E. Matijevic, “Formation of uniform spherical magnetite particles by crystallization from ferrous hydroxide gels,” Journal of Colloid and Interface Science, vol. 74, no. 1, pp. 187–193, 1980.
[44]
R. Massart, “Preparation of aqueous magnetic liquids in alkaline and acidic media,” IEEE Transactions on Magnetics, vol. 17, no. 2, pp. 1247–1248, 1981.
[45]
S. Neveu, A. Bee, M. Robineau, and D. Talbot, “Size-selective chemical synthesis of tartrate stabilized cobalt ferrite ionic magnetic fluid,” Journal of Colloid and Interface Science, vol. 255, no. 2, pp. 293–298, 2002.
[46]
J. Cheon, N.-J. Kang, S.-M. Lee, J.-H. Lee, J.-H. Yoon, and S. J. Oh, “Shape evolution of single-crystalline iron oxide nanocrystals,” Journal of the American Chemical Society, vol. 126, no. 7, pp. 1950–1951, 2004.
[47]
Y.-W. Jun, J.-S. Choi, and J. Cheon, “Heterostructured magnetic nanoparticles: their versatility and high performance capabilities,” Chemical Communications, no. 12, pp. 1203–1214, 2007.
[48]
M. F. Dumont, O. N. Risset, E. S. Knowles et al., “Synthesis and size control of iron(II) hexacyanochromate(III) nanoparticles and the effect of particle size on linkage isomerism,” Inorganic Chemistry, vol. 52, no. 8, pp. 4494–4501, 2013.
[49]
C. Aubery, C. Solans, S. Prevost, M. Gradzielski, and M. Sanchez-Dominguez, “Microemulsions as reaction media for the synthesis of mixed oxide nanoparticles: relationships between microemulsion structure, reactivity, and nanoparticle characteristics,” Langmuir, vol. 29, no. 6, pp. 1779–1789, 2013.
[50]
C. Okoli, M. Sanchez-Dominguez, M. Boutonnet et al., “Comparison and functionalization study of microemulsion-prepared magnetic iron oxide nanoparticles,” Langmuir, vol. 28, no. 22, pp. 8479–8485, 2012.
[51]
S. Chkoundali, S. Ammar, N. Jouini et al., “Nickel ferrite nanoparticles: elaboration in polyol medium via hydrolysis, and magnetic properties,” Journal of Physics: Condensed Matter, vol. 16, no. 24, article 4357, 2004.
[52]
Y. F. Shen, J. Tang, Z. H. Nie, Y. D. Wang, Y. Ren, and L. Zuo, “Tailoring size and structural distortion of Fe3O4 nanoparticles for the purification of contaminated water,” Bioresource Technology, vol. 100, no. 18, pp. 4139–4146, 2009.
[53]
S. Sun, H. Zeng, D. B. Robinson et al., “Monodisperse MFe2O4 (M = Fe, Co, Mn) Nanoparticles,” Journal of the American Chemical Society, vol. 126, no. 1, pp. 273–279, 2004.
[54]
S. K. Choi, A. Myc, J. E. Silpe et al., “Dendrimer-based multivalent vancomycin nanoplatform for targeting the drug-resistant bacterial surface,” ACS Nano, vol. 7, no. 1, pp. 214–228, 2013.
[55]
A.-L. Morel, S. I. Nikitenko, K. Gionnet et al., “Sonochemical approach to the synthesis of Fe3O4 @SiO2 core—shell nanoparticles with tunable properties,” ACS Nano, vol. 2, no. 5, pp. 847–856, 2008.
[56]
F. Dang, N. Enomoto, J. Hojo, and K. Enpuku, “Sonochemical synthesis of monodispersed magnetite nanoparticles by using an ethanol-water mixed solvent,” Ultrasonics Sonochemistry, vol. 16, no. 5, pp. 649–654, 2009.
[57]
Y. Xue, H. Chen, D. Yu et al., “Oxidizing metal ions with graphene oxide: the in situ formation of magnetic nanoparticles on self-reduced graphene sheets for multifunctional applications,” Chemical Communications, vol. 47, no. 42, pp. 11689–11691, 2011.
[58]
D. Grumelli, C. Vericat, G. Benítez et al., “Electrochemical preparation and delivery of melanin-iron covered gold nanoparticles,” ChemPhysChem, vol. 10, no. 2, pp. 370–373, 2009.
[59]
T. Rudin, K. Wegner, and S. E. Pratsinis, “Uniform nanoparticles by flame-assisted spray pyrolysis (FASP) of low cost precursors,” Journal of Nanoparticle Research, vol. 13, no. 7, pp. 2715–2725, 2011.
[60]
W. Y. Teoh, R. Amal, and L. M?dler, “Flame spray pyrolysis: an enabling technology for nanoparticles design and fabrication,” Nanoscale, vol. 2, no. 8, pp. 1324–1347, 2010.
[61]
O. Bomati-Miguel, P. Tartaj, M. P. Morales et al., “Core-shell iron-iron oxide nanoparticles synthesized by laser-induced pyrolysis,” Small, vol. 2, no. 12, pp. 1476–1483, 2006.
[62]
I. Morjan, R. Alexandrescu, F. Dumitrache et al., “Iron Oxide-Based nanoparticles with different mean sizes obtained by the laser pyrolysis: structural and magnetic properties,” Journal of Nanoscience and Nanotechnology, vol. 10, no. 2, pp. 1223–1234, 2010.
[63]
C. Liu, P. Zhang, X. Zhai et al., “Nano-carrier for gene delivery and bioimaging based on carbon dots with PEI-passivation enhanced fluorescence,” Biomaterials, vol. 33, no. 13, pp. 3604–3613, 2012.
[64]
B. Koppolu, Z. Bhavsar, A. S. Wadajkar et al., “Temperature-sensitive polymer-coated magnetic nanoparticles as a potential drug delivery system for targeted therapy of thyroid cancer,” Journal of Biomedical Nanotechnology, vol. 8, no. 6, pp. 983–990, 2012.
[65]
K. Katagiri, Y. Imai, K. Koumoto, T. Kaiden, K. Kono, and S. Aoshima, “Magnetoresponsive on-demand release of hybrid liposomes formed from Fe3O4 nanoparticles and thermosensitive block copolymers,” Small, vol. 7, no. 12, pp. 1683–1689, 2011.
[66]
S. S. Yu, R. L. Scherer, R. A. Ortega et al., “Enzymatic- and temperature-sensitive controlled release of ultrasmall superparamagnetic iron oxides (USPIOs),” Journal of Nanobiotechnology, vol. 9, no. 1, article 7, 2011.
[67]
T.-Y. Liu, S.-H. Hu, K.-H. Liu, R.-S. Shaiu, D.-M. Liu, and S.-Y. Chen, “Instantaneous drug delivery of magnetic/thermally sensitive nanospheres by a high-frequency magnetic field,” Langmuir, vol. 24, no. 23, pp. 13306–13311, 2008.
[68]
G. Mikhaylov, U. Mikac, A. A. Magaeva et al., “Ferri-liposomes as an MRI-visible drug-delivery system for targeting tumours and their microenvironment,” Nature Nanotechnology, vol. 6, no. 9, pp. 594–602, 2011.
[69]
B. Clares, R. A. Biedma-Ortiz, E. Sáez-Fernández et al., “Nano-engineering of 5-fluorouracil-loaded magnetoliposomes for combined hyperthermia and chemotherapy against colon cancer,” European Journal of Pharmaceutics and Biopharmaceutics, 2013.
[70]
M. R. Faria, M. M. Cruz, M. C. Gon?alves, A. Carvalho, G. Feio, and M. B. F. Martins, “Synthesis and characterization of magnetoliposomes for MRI contrast enhancement,” International Journal of Pharmaceutics, vol. 446, no. 1-2, pp. 183–190, 2013.
[71]
D. Qiu and X. An, “Controllable release from magnetoliposomes by magnetic stimulation and thermal stimulation,” Colloids Surfaces B, vol. 104, pp. 326–329, 2013.
[72]
S. García-Jimeno, E. Escribano, J. Queralt, and J. Estelrich, “External magnetic field-induced selective biodistribution of magnetoliposomes in mice,” Nanoscale Research Letters, vol. 7, no. 1, article 452, 2012.
[73]
B. Garnier, S. Tan, S. Miraux, E. Bled, and A. R. Brisson, “Optimized synthesis of 100?nm diameter magnetoliposomes with high content of maghemite particles and high MRI effect,” Contrast Media and Molecular Imaging, vol. 7, no. 2, pp. 231–239, 2012.
[74]
J. Hanu?, M. Ullrich, J. Dohnal, M. Singh, and F. Stěpánek, “Remotely controlled diffusion from magnetic liposome microgels,” Langmuir, vol. 29, no. 13, pp. 4381–4387, 2013.
[75]
L. Deng, X. Ke, Z. He et al., “A MSLN-targeted multifunctional nanoimmunoliposome for MRI and targeting therapy in pancreatic cancer,” International Journal of Nanomedicine, vol. 7, pp. 5053–5065, 2012.
[76]
R. Mejías, R. Costo, A. G. Roca et al., “Cytokine adsorption/release on uniform magnetic nanoparticles for localized drug delivery,” Journal of Controlled Release, vol. 130, no. 2, pp. 168–174, 2008.
[77]
R. Mejías, S. Pérez-Yagüe, A. G. Roca et al., “Liver and brain imaging through dimercaptosuccinic acid-coated iron oxide nanoparticles,” Nanomedicine, vol. 5, no. 3, pp. 397–408, 2010.
[78]
R. Mejías, S. Pérez-Yagüe, L. Gutiérrez et al., “Dimercaptosuccinic acid-coated magnetite nanoparticles for magnetically guided in vivo delivery of interferon gamma for cancer immunotherapy,” Biomaterials, vol. 32, no. 11, pp. 2938–2952, 2011.
[79]
Y. Gu, P. Song, T. Li, and W. Sui, “Synthesis and characterization of carboxymethyl-polyaminate chitosan and its adsorption behavior toward a reactive dye,” Carbohydrate Research, vol. 346, no. 6, pp. 769–774, 2011.
[80]
H. Lee, K. Y. Mi, S. Park et al., “Thermally cross-linked superparamagnetic iron oxide nanoparticles: synthesis and application as a dual imaging probe for cancer in vivo,” Journal of the American Chemical Society, vol. 129, no. 42, pp. 12739–12745, 2007.
[81]
J. Xie, K. Chen, H.-Y. Lee et al., “Ultrasmall c(RGDyK)-coated Fe3O4 nanoparticles and their specific targeting to integrin αvβ3-rich tumor cells,” Journal of the American Chemical Society, vol. 130, no. 24, pp. 7542–7543, 2008.
[82]
F. Bettio, M. Canevari, C. Marzano et al., “Synthesis and biological in vitro evaluation of novel PEG-psoralen conjugates,” Biomacromolecules, vol. 7, no. 12, pp. 3534–3541, 2006.
[83]
L. Yuan, W. Chen, J. Hu, J. Z. Zhang, and D. Yang, “Mechanistic study of the covalent loading of paclitaxel via disulfide linkers for controlled drug release,” Langmuir, vol. 29, no. 2, pp. 734–743, 2013.
[84]
J. K. Eby, K. Y. Dane, C. P. O'Neil, S. Hirosue, M. A. Swartz, and J. Hubbell, “Polymer micelles with pyridyl disulfide-coupled antigen travel through lymphatics and show enhanced cellular responses following immunization,” Acta Biomaterialia, vol. 8, no. 9, pp. 3210–3217, 2012.
[85]
W. Xiao, J. Lin, M. Li et al., “Prolonged in vivo circulation time by zwitterionic modification of magnetite nanoparticles for blood pool contrast agents,” Contrast Media and Molecular Imaging, vol. 7, no. 3, pp. 320–327, 2012.
[86]
J. Guo, X. Gao, L. Su et al., “Aptamer-functionalized PEG-PLGA nanoparticles for enhanced anti-glioma drug delivery,” Biomaterials, vol. 32, no. 31, pp. 8010–8020, 2011.
[87]
H. Ando, A. Okamoto, M. Yokota et al., “Development of miR-92a delivery system for antiangiogenesis-based cancer therapy,” The Journal of Gene Medicine, vol. 15, no. 1, pp. 20–27, 2012.
[88]
S. K. Tripathi, R. Goyal, P. Kumar, and K. C. Gupta, “Linear polyethylenimine-graft-chitosan copolymers as efficient DNA/siRNA delivery vectors in vitro and in vivo,” Nanomedicine, vol. 8, no. 3, pp. 337–345, 2012.
[89]
Y. Bai, F. Feng, L. Zhao et al., “Aptamer/thrombin/aptamer-AuNPs sandwich enhanced surface plasmon resonance sensor for the detection of subnanomolar thrombin,” Biosensors Bioelectronics, vol. 47, pp. 265–270, 2013.
[90]
W. G. Lewis, L. G. Green, F. Grynszpan et al., “Click chemistry in situ: acetylcholinesterase as a reaction vessel for the selective assembly of a femtomolar inhibitor from an array of building blocks,” Angewandte Chemie, vol. 41, no. 6, pp. 1053–1057, 2002.
[91]
G. Von Maltzahn, Y. Ren, J.-H. Park et al., “In vivo tumor cell targeting with “click” nanoparticles,” Bioconjugate Chemistry, vol. 19, no. 8, pp. 1570–1578, 2008.
[92]
T. C. Chu, F. Shieh, L. A. Lavery et al., “Labeling tumor cells with fluorescent nanocrystal-aptamer bioconjugates,” Biosensors and Bioelectronics, vol. 21, no. 10, pp. 1859–1866, 2006.
[93]
D. J. Javier, N. Nitin, M. Levy, A. Ellington, and R. Richards-Kortum, “Aptamer-targeted gold nanoparticles as molecular-specific contrast agents for reflectance imaging,” Bioconjugate Chemistry, vol. 19, no. 6, pp. 1309–1312, 2008.
[94]
W. Yang, “Nucleases: diversity of structure, function and mechanism,” Quarterly Reviews of Biophysics, vol. 44, no. 1, pp. 1–93, 2011.
[95]
F. Liu, L. M. Shollenberger, C. C. Conwell, X. Yuan, and L. Huang, “Mechanism of naked DNA clearance after intravenous injection,” Journal of Gene Medicine, vol. 9, no. 7, pp. 613–619, 2007.
[96]
S. T. Sarvestani, B. R. G. Williams, and M. P. Gantier, “Human Toll-like receptor 8 can be cool too: implications for foreign RNA sensing,” Journal of Interferon & Cytokine Research, vol. 32, no. 8, pp. 350–361, 2012.
[97]
V. Gosu, S. Basith, O. P. Kwon, and S. Choi, “Therapeutic applications of nucleic acids and their analogues in Toll-like receptor signaling,” Molecules, vol. 17, no. 11, pp. 13503–13529, 2012.
[98]
H. Aldawsari, B. S. Raj, R. Edrada-Ebel et al., “Enhanced gene expression in tumors after intravenous administration of arginine-, lysine- and leucine-bearing polyethylenimine polyplex,” Nanomedicine, vol. 7, no. 5, pp. 615–623, 2011.
[99]
C. Chandrashekhar, B. Pons, C. D. Muller, N. Tounsi, R. Mulherkar, and G. Zuber, “Oligobenzylethylenimine enriches linear polyethylenimine with a pH-sensitive membrane-disruptive property and leads to enhanced gene delivery activity,” Acta Biomaterialia, vol. 9, no. 2, pp. 4985–4993, 2012.
[100]
W. Fan, X. Wu, B. Ding et al., “Degradable gene delivery systems based on Pluronics-modified low-molecular-weight polyethylenimine: preparation, characterization, intracellular trafficking, and cellular distribution,” International Journal of Nanomedicine, vol. 7, pp. 1127–1138, 2012.
[101]
R. Goyal, S. K. Tripathi, S. Tyagi et al., “Linear PEI nanoparticles: efficient pDNA/siRNA carriers in vitro and in vivo,” Nanomedicine, vol. 8, no. 2, pp. 167–175, 2012.
[102]
A. Kwok and S. L. Hart, “Comparative structural and functional studies of nanoparticle formulations for DNA and siRNA delivery,” Nanomedicine, vol. 7, no. 2, pp. 210–219, 2011.
[103]
M. Malhotra, C. Tomaro-Duchesneau, and S. Prakash, “Synthesis of TAT peptide-tagged PEGylated chitosan nanoparticles for siRNA delivery targeting neurodegenerative diseases,” Biomaterials, vol. 34, no. 4, pp. 1270–1280, 2012.
[104]
C. Liu, F. Liu, L. Feng, M. Li, J. Zhang, and N. Zhang, “The targeted co-delivery of DNA and doxorubicin to tumor cells via multifunctional PEI-PEG based nanoparticles,” Biomaterials, vol. 34, no. 10, pp. 2547–2564, 2013.
[105]
Y. He, G. Cheng, L. Xie, Y. Nie, B. He, and Z. Gu, “Polyethyleneimine/DNA polyplexes with reduction-sensitive hyaluronic acid derivatives shielding for targeted gene delivery,” Biomaterials, vol. 34, no. 4, pp. 1235–1245, 2012.
[106]
R. K. Oskuee, A. Dehshahri, W. T. Shier, and M. Ramezani, “Alkylcarboxylate grafting to polyethylenimine: a simple approach to producing a DNA nanocarrier with low toxicity,” Journal of Gene Medicine, vol. 11, no. 10, pp. 921–932, 2009.
[107]
S. Patnaik, M. Arif, A. Pathak, R. Kurupati, Y. Singh, and K. C. Gupta, “Cross-linked polyethylenimine-hexametaphosphate nanoparticles to deliver nucleic acids therapeutics,” Nanomedicine, vol. 6, no. 2, pp. 344–354, 2010.
[108]
M. Garcia-Fuentes and M. J. Alonso, “Chitosan-based drug nanocarriers: where do we stand?” Journal of Controlled Release, vol. 161, no. 2, pp. 496–504, 2012.
[109]
E. K.-W. Toh, H.-Y. Chen, Y.-L. Lo, S.-J. Huang, and L.-F. Wang, “Succinated chitosan as a gene carrier for improved chitosan solubility and gene transfection,” Nanomedicine, vol. 7, no. 2, pp. 174–183, 2011.
[110]
J. H. Na, S. Y. Lee, S. Lee et al., “Effect of the stability and deformability of self-assembled glycol chitosan nanoparticles on tumor-targeting efficiency,” Journal of Controlled Release, vol. 163, no. 1, pp. 2–9, 2012.
[111]
J. H. Na, H. Koo, S. Lee et al., “Real-time and non-invasive optical imaging of tumor-targeting glycol chitosan nanoparticles in various tumor models,” Biomaterials, vol. 32, no. 22, pp. 5252–5261, 2011.
[112]
R. Reul, J. Nguyen, A. Biela et al., “Biophysical and biological investigation of DNA nano-complexes with a non-toxic, biodegradable amine-modified hyperbranched polyester,” International Journal of Pharmaceutics, vol. 436, no. 1-2, pp. 97–105, 2012.
[113]
M. Monaghan, U. Greiser, H. Cao, W. Wang, and A. Pandit, “An antibody fragment functionalized dendritic PEGylated poly(2-(dimethylamino)ethyl diacrylate) as a vehicle of exogenous microRNA,” Drug Delivery and Translational Research, vol. 2, no. 5, pp. 406–414, 2012.
[114]
T. Merdan, J. Callahan, H. Petersen et al., “Pegylated polyethylenimine-Fab′ antibody fragment conjugates for targeted gene delivery to human ovarian carcinoma cells,” Bioconjugate Chemistry, vol. 14, no. 5, pp. 989–996, 2003.
[115]
Y. Chen, W. Wang, G. Lian et al., “Development of an MRI-visible nonviral vector for siRNA delivery targeting gastric cancer,” International Journal of Nanomedicine, vol. 7, pp. 359–368, 2012.
[116]
K. K. Hou, H. Pan, G. M. Lanza, and S. A. Wickline, “Melittin derived peptides for nanoparticle based siRNA transfection,” Biomaterials, vol. 34, no. 12, pp. 3110–3119, 2013.
[117]
G. D. Kenny, C. Villegas-Llerena, A. D. Tagalakis et al., “Multifunctional receptor-targeted nanocomplexes for magnetic resonance imaging and transfection of tumours,” Biomaterials, vol. 33, no. 29, pp. 7241–7250, 2012.
[118]
H. Y. Wang, W. J. Yi, S. Y. Qin, C. Li, R. X. Zhuo, and X. Z. Zhang, “Tyroserleutide-based gene vector for suppressing VEGF expression in cancer therapy,” Biomaterials, vol. 33, no. 33, pp. 8685–8694, 2012.
[119]
M. K. Yu, J. Park, and S. Jon, “Targeting strategies for multifunctional nanoparticles in cancer imaging and therapy,” Theranostics, vol. 2, no. 1, pp. 3–44, 2012.
[120]
F. Kr?tz, C. de Wit, H.-Y. Sohn et al., “Magnetofection—a highly efficient tool for antisense oligonucleotide delivery in vitro and in vivo,” Molecular Therapy, vol. 7, no. 5, pp. 700–710, 2003.
[121]
C. Plank, F. Scherer, U. Schillinger, C. Bergemann, and M. Anton, “Magnetofection: enhancing and targeting gene delivery with superparamagnetic nanoparticles and magnetic fields,” Journal of Liposome Research, vol. 13, no. 1, pp. 29–32, 2003.
[122]
Y. Chen, G. Lian, C. Liao et al., “Characterization of polyethylene glycol-grafted polyethylenimine and superparamagnetic iron oxide nanoparticles (PEG-g-PEI-SPION) as an MRI-visible vector for siRNA delivery in gastric cancer in vitro and in vivo,” Journal of Gastroenterology, vol. 48, no. 7, pp. 809–821, 2013.
[123]
R. Namgung, K. Singha, M. K. Yu et al., “Hybrid superparamagnetic iron oxide nanoparticle-branched polyethylenimine magnetoplexes for gene transfection of vascular endothelial cells,” Biomaterials, vol. 31, no. 14, pp. 4204–4213, 2010.
[124]
M. Shen, F. Gong, P. Pang et al., “An MRI-visible non-viral vector for targeted Bcl-2 siRNA delivery to neuroblastoma,” International Journal of Nanomedicine, vol. 7, pp. 3319–3332, 2012.
[125]
H. J. Lee, Y. T. C. Nguyen, M. Muthiah et al., “MR traceable delivery of p53 tumor suppressor gene by PEI-functionalized superparamagnetic iron oxide nanoparticles,” Journal of Biomedical Nanotechnology, vol. 8, no. 3, pp. 361–371, 2012.
[126]
G. Liu, J. Xie, F. Zhang et al., “N-Alkyl-PEI-functionalized iron oxide nanoclusters for efficient siRNA delivery,” Small, vol. 7, no. 19, pp. 2742–2749, 2011.
[127]
L. Zhang, T. Wang, L. Li, C. Wang, Z. Su, and J. Li, “Multifunctional fluorescent-magnetic polyethyleneimine functionalized Fe3O4-mesoporous silica yolk-shell nanocapsules for siRNA delivery,” Chemical Communications, vol. 48, no. 69, pp. 8706–8708, 2012.
[128]
L. Gao, L. Xie, X. Long et al., “Efficacy of MRI visible iron oxide nanoparticles in delivering minicircle DNA into liver via intrabiliary infusion,” Biomaterials, vol. 34, no. 14, pp. 3688–3696, 2013.
[129]
L. Qi, L. Wu, S. Zheng, Y. Wang, H. Fu, and D. Cui, “Cell-penetrating magnetic nanoparticles for highly efficient delivery and intracellular imaging of siRNA,” Biomacromolecules, vol. 13, no. 9, pp. 2723–2730, 2012.
[130]
L. Miao, K. Zhang, C. Qiao et al., “Antitumor effect of human TRAIL on adenoid cystic carcinoma using magnetic nanoparticle-mediated gene expression,” Nanomedicine, vol. 9, no. 1, pp. 141–150, 2013.
[131]
J. Qian, A. Dong, M. Kong, Z. Ma, J. Fan, and G. Jiang, “Suppression of type 1 insulin-like growth factor receptor expression by small interfering RNA inhibits A549 human lung cancer cell invasion in vitro and metastasis in xenograft nude mice,” Acta Biochimica et Biophysica Sinica, vol. 39, no. 2, pp. 137–147, 2007.
[132]
A.-Q. Dong, M.-J. Kong, Z.-Y. Ma, J.-F. Qian, and X.-H. Xu, “Down-regulation of IGF-IR using small, interfering, hairpin RNA (siRNA) inhibits growth of human lung cancer cell line A549 in vitro and in nude mice,” Cell Biology International, vol. 31, no. 5, pp. 500–507, 2007.
[133]
A. Dong, M. Kong, Z. Ma, J. Qian, H. Cheng, and X. Xu, “Knockdown of insulin-like growth factor 1 receptor enhances chemosensitivity to cisplatin in human lung adenocarcinoma A549 cells,” Acta Biochimica et Biophysica Sinica, vol. 40, no. 6, pp. 497–504, 2008.
[134]
C. Wang, C. Ding, M. Kong et al., “Tumor-targeting magnetic lipoplex delivery of short hairpin RNA suppresses IGF-1R overexpression of lung adenocarcinoma A549 cells in vitro and in vivo,” Biochemical and Biophysical Research Communications, vol. 410, no. 3, pp. 537–542, 2011.
[135]
M. Kong, X. Li, C. Wang et al., “Tissue distribution and cancer growth inhibition of magnetic lipoplex-delivered type 1 insulin-like growth factor receptor shRNA in nude mice,” Acta Biochimica et Biophysica Sinica, vol. 44, pp. 591–596, 2012.
[136]
H. Mannell, J. Pircher, F. Fochler et al., “Site directed vascular gene delivery in vivo by ultrasonic destruction of magnetic nanoparticle coated microbubbles,” Nanomedicine, vol. 8, no. 8, pp. 1309–1318, 2012.
[137]
S. Jiang, A. A. Eltoukhy, K. T. Love, R. Langer, and D. G. Anderson, “Lipidoid-coated iron oxide nanoparticles for efficient DNA and siRNA delivery,” NanoLetters, vol. 13, no. 3, pp. 1059–1064, 2013.
[138]
H. Mok, O. Veiseh, C. Fang et al., “PH-sensitive siRNA nanovector for targeted gene silencing and cytotoxic effect in cancer cells,” Molecular Pharmaceutics, vol. 7, no. 6, pp. 1930–1939, 2010.
[139]
Y. Namiki, T. Namiki, H. Yoshida et al., “A novel magnetic crystal-lipid nanostructure for magnetically guided in vivo gene delivery,” Nature Nanotechnology, vol. 4, no. 9, pp. 598–606, 2009.
[140]
S.-L. Sun, Y.-L. Lo, H.-Y. Chen, and L.-F. Wang, “Hybrid polyethylenimine and polyacrylic acid-bound iron oxide as a magnetoplex for gene delivery,” Langmuir, vol. 28, no. 7, pp. 3542–3552, 2012.
[141]
J.-H. Hwang, S. Lee, E. Kim et al., “Heparin-coated superparamagnetic nanoparticle-mediated adeno-associated virus delivery for enhancing cellular transduction,” International Journal of Pharmaceutics, vol. 421, no. 2, pp. 397–404, 2011.
[142]
E. Kim, J.-S. Oh, I.-S. Ahn, K. I. Park, and J.-H. Jang, “Magnetically enhanced adeno-associated viral vector delivery for human neural stem cell infection,” Biomaterials, vol. 32, no. 33, pp. 8654–8662, 2011.
[143]
C. Sapet, C. Pellegrino, N. Laurent, F. Sicard, and O. Zelphati, “Magnetic nanoparticles enhance adenovirus transduction in vitro and in vivo,” Pharmaceutical Research, vol. 29, no. 5, pp. 1203–1218, 2012.