425
Views
28
CrossRef citations to date
0
Altmetric
Review

Non-viral nucleic acid containing nanoparticles as cancer therapeutics

, &
Pages 1475-1487 | Received 06 Mar 2016, Accepted 13 May 2016, Published online: 06 Jun 2016

References

  • Manley JL, Sharp PA, Gefter ML. RNA synthesis in isolated nuclei: in vitro initiation of adenovirus 2 major late mRNA precursor. Proc Natl Acad Sci. 1979;76:160–164.
  • Manthorpe M, Cornefert-Jensen F, Hartikka J, et al. Gene therapy by intramuscular injection of plasmid DNA: studies on firefly luciferase gene expression in mice. Hum Gene Ther. 1993;4:419–431.
  • Liu F, Song Y, Liu D. Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA. Gene Ther. 1999;6:1258–1266.
  • Fire A, Xu SQ, Montgomery MK, et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391:806–811.
  • Bernstein E, Caudy AA, Hammond SM, et al. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature. 2001;409:363–366.
  • Hannon GJ. RNA interference. Nature. 2002;418:244–251.
  • Hickman MA, Malone RW, Lehmann-Bruinsma K, et al. Gene expression following direct injection of DNA into liver. Hum Gene Ther. 1994;5:1477–1483.
  • Clemens JC, Worby CA, Simonson-Leff N, et al. Use of double-stranded RNA interference in Drosophila cell lines to dissect signal transduction pathways. Proc Natl Acad Sci U S A. 2000;97:6499–6503.
  • Rosenfeld MA, Siegfried W, Yoshimura K, et al. Adenovirus-mediated transfer of a recombinant α1-antitrypsin gene to the lung epithelium in vivo. Science. 1991;252:431–434.
  • Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet. 2003;4:346–358.
  • Pack DW, Hoffman AS, Pun S, et al. Design and development of polymers for gene delivery. Nat Rev Drug Discov. 2005;4:581–593.
  • Alving CR, Steck EA, Chapman WL Jr., et al. Therapy of leishmaniasis: superior efficacies of liposome-encapsulated drugs. Proc Natl Acad Sci. 1978;75:2959–2963.
  • Vadiei K, Lopez-Berestein G, Perez-Soler R, et al. In vitro evaluation of liposomal cyclosporine. Int J Pharm. 1989;57:133–138.
  • Scherphof GL, Dijkstra J, Spanjer HH, et al. Uptake and intracellular processing of targeted and nontargeted liposomes by rat Kupffer cells in vivo and in vitro. Ann N Y Acad Sci. 1985;446:368–384.
  • Wagner E, Plank C, Zatloukal K, et al. Influenza-virus hemagglutinin-Ha-2 N-terminal fusogenic peptides augment gene-transfer by transferrin polylysine DNA complexes - toward a synthetic virus-like gene-transfer vehicle. Proc Natl Acad Sci. 1992;89:7934–7938.
  • Wu GY, Wu CH. Receptor-mediated in vitro gene transformation by a soluble DNA carrier system. J Biol Chem. 1987;262:4429–4432.
  • Giljohann DA, Seferos DS, Prigodich AE, et al. Gene regulation with polyvalent siRNA-nanoparticle conjugates. J Am Chem Soc. 2009;131:2072–2073.
  • Hagerman PJ. Flexibility of RNA. Annu Rev Bioph Biom. 1997;26:139–156.
  • Kebbekus P, Draper DE, Hagerman P. Persistence length of RNA. Biochemistry-Us. 1995;34:4354–4357.
  • Ogris M, Brunner S, Schuller S, et al. PEGylated DNA/transferrin-PEI complexes: reduced interaction with blood components, extended circulation in blood and potential for systemic gene delivery. Gene Ther. 1999;6:595–605.
  • Frankel AD, Pabo CO. Cellular uptake of the tat protein from human immunodeficiency virus. Cell. 1988;55:1189–1193.
  • Green M, Loewenstein PM. Autonomous functional domains of chemically synthesized human immunodeficiency virus tat trans-activator protein. Cell. 1988;55:1179–1188.
  • Wyman TB, Nicol F, Zelphati O, et al. Design, synthesis, and characterization of a cationic peptide that binds to nucleic acids and permeabilizes bilayers. Biochemistry-Us. 1997;36:3008–3017.
  • Hafez I, Maurer N, Cullis P. On the mechanism whereby cationic lipids promote intracellular delivery of polynucleic acids. Gene Ther. 2001;8:1188–1196.
  • Xu Y, Szoka FC Jr. Mechanism of DNA release from cationic liposome/DNA complexes used in cell transfection. Biochemistry-Us. 1996;35:5616–5623.
  • Zelphati O, Szoka FC Jr. Mechanism of oligonucleotide release from cationic liposomes. Proc Natl Acad Sci. 1996;93:11493–11498.
  • Verma A, Stellacci F. Effect of surface properties on nanoparticle-cell interactions. Small. 2010;6:12–21.
  • Felgner JH, Kumar R, Sridhar CN, et al. Enhanced gene delivery and mechanism studies with a novel series of cationic lipid formulations. J Biol Chem. 1994;269:2550–2561.
  • El Ouahabi A, Thiry M, Pector V, et al. The role of endosome destabilizing activity in the gene transfer process mediated by cationic lipids. Febs Lett. 1997;414:187–192.
  • Benjaminsen RV, Mattebjerg MA, Henriksen JR, et al. The possible “Proton Sponge” effect of polyethylenimine (PEI) does not include change in lysosomal pH. Mol Therapy. 2013;21:149–157.
  • Sonawane ND, Szoka FC, Verkman AS. Chloride accumulation and swelling in endosomes enhances DNA transfer by polyamine-DNA polyplexes. J Biol Chem. 2003;278:44826–44831.
  • Nel AE, Madler L, Velegol D, et al. Understanding biophysicochemical interactions at the nano-bio interface. Nat Mater. 2009;8:543–557.
  • Kawasaki H, Taira K. Short hairpin type of dsRNAs that are controlled by tRNAVal promoter significantly induce RNAi-mediated gene silencing in the cytoplasm of human cells. Nucleic Acids Res. 2003;31:700–707.
  • Lynn DM, Langer R. Degradable poly(β-amino esters): synthesis, characterization, and self-assembly with plasmid DNA. J Am Chem Soc. 2000;122:10761–10768.
  • Woodrow KA, Cu Y, Booth CJ, et al. Intravaginal gene silencing using biodegradable polymer nanoparticles densely loaded with small-interfering RNA. Nat Mater. 2009;8:526–533.
  • Griffith OW. Biologic and pharmacologic regulation of mammalian glutathione synthesis. Free Radic Biol Med. 1999;27:922–935.
  • Son S, Namgung R, Kim J, et al. Bioreducible polymers for gene silencing and delivery. Acc Chem Res. 2012;45:1100–1112.
  • Lukacs GL, Haggie P, Seksek O, et al. Size-dependent DNA mobility in cytoplasm and nucleus. J Biol Chem. 2000;275:1625–1629.
  • Lechardeur D, Sohn KJ, Haardt M, et al. Metabolic instability of plasmid DNA in the cytosol: a potential barrier to gene transfer. Gene Ther. 1999;6:482–497.
  • Brunner S, Sauer T, Carotta S, et al. Cell cycle dependence of gene transfer by lipoplex, polyplex and recombinant adenovirus. Gene Ther. 2000;7:401–407.
  • Zanta MA, Belguise-Valladier P, Behr J-P. Gene delivery: a single nuclear localization signal peptide is sufficient to carry DNA to the cell nucleus. Proc Natl Acad Sci. 1999;96:91–96.
  • Grandinetti G, Reineke TM. Exploring the mechanism of plasmid DNA nuclear internalization with polymer-based vehicles. Mol Pharm. 2012;9:2256–2267.
  • Graham FL, Van Der Eb AJ. A new technique for the assay of infectivity of human adenovirus 5 DNA. Virology. 1973;52:456–467.
  • Kester M, Heakal Y, Fox T, et al. Calcium phosphate nanocomposite particles for in vitro imaging and encapsulated chemotherapeutic drug delivery to cancer cells. Nano Lett. 2008;8:4116–4121.
  • Liu T, Tang A, Zhang G, et al. Calcium phosphate nanoparticles as a novel nonviral vector for efficient transfection of DNA in cancer gene therapy. Cancer Biother Radiopharm. 2005;20:141–149.
  • Fraley R, Subramani S, Berg P, et al. Introduction of liposome-encapsulated SV40 DNA into cells. J Biol Chem. 1980;255:10431–10435.
  • Campbell RB, Fukumura D, Brown EB, et al. Cationic charge determines the distribution of liposomes between the vascular and extravascular compartments of tumors. Cancer Res. 2002;62:6831–6836.
  • Thurston G, McLean JW, Rizen M, et al. Cationic liposomes target angiogenic endothelial cells in tumors and chronic inflammation in mice. J Clin Invest. 1998;101:1401.
  • Torchilin VP, Levchenko TS, Rammohan R, et al. Cell transfection in vitro and in vivo with nontoxic TAT peptide-liposome–DNA complexes. Proc Natl Acad Sci. 2003;100:1972–1977.
  • Cristiano RJ, Roth JA. Epidermal growth factor mediated DNA delivery into lung cancer cells via the epidermal growth factor receptor. Cancer Gene Ther. 1995;3:4–10.
  • Tanaka M, Fraizer GC, De La Cerda J, et al. Connexin 26 enhances the bystander effect in HSVtk/GCV gene therapy for human bladder cancer by adenovirus/PLL/DNA gene delivery. Gene Ther. 2001;8:139–148.
  • Yang NS, Burkholder J, Roberts B, et al. In vivo and in vitro gene transfer to mammalian somatic cells by particle bombardment. Proc Natl Acad Sci. 1990;87:9568–9572.
  • Visaria RK, Griffin RJ, Williams BW, et al. Enhancement of tumor thermal therapy using gold nanoparticle–assisted tumor necrosis factor-α delivery. Mol Cancer Ther. 2006;5:1014–1020.
  • Wang F, Wang Y-C, Dou S, et al. Doxorubicin-tethered responsive gold nanoparticles facilitate intracellular drug delivery for overcoming multidrug resistance in cancer cells. ACS Nano. 2011;5:3679–3692.
  • Haensler J, Szoka FC. Polyamidoamine cascade polymers mediate efficient transfection of cells in culture. Bioconjug Chem. 1993;4:372–379.
  • Loup C, Zanta MA, Caminade AM, et al. Preparation of water‐soluble cationic phosphorus‐containing dendrimers as DNA transfecting agents. Chemistry–A Eur J. 1999;5:3644–3650.
  • Wang P, Zhao X-H, Wang Z-Y, et al. Generation 4 polyamidoamine dendrimers is a novel candidate of nano-carrier for gene delivery agents in breast cancer treatment. Cancer Lett. 2010;298:34–49.
  • Patil ML, Zhang M, Taratula O, et al. Internally cationic polyamidoamine PAMAM-OH Dendrimers for sirna delivery: effect of the degree of quaternization and cancer targeting. Biomacromolecules. 2009;10:258–266.
  • Koppu S, Oh YJ, Edrada-Ebel R, et al. Tumor regression after systemic administration of a novel tumor-targeted gene delivery system carrying a therapeutic plasmid DNA. J Control Release. 2010;143:215–221.
  • Boussif O, Lezoualc’h F, Zanta MA, et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci U S A. 1995;92:7297–7301.
  • Schiffelers RM, Ansari A, Xu J, et al. Cancer siRNA therapy by tumor selective delivery with ligand-targeted sterically stabilized nanoparticle. Nucleic Acids Res. 2004;32:e149–e.
  • Veiseh O, Kievit FM, Gunn JW, et al. A ligand-mediated nanovector for targeted gene delivery and transfection in cancer cells. Biomaterials. 2009;30:649–657.
  • Liang B, He M-L, Xiao Z-P, et al. Synthesis and characterization of folate-PEG-grafted-hyperbranched-PEI for tumor-targeted gene delivery. Biochem Biophys Res Commun. 2008;367:874–880.
  • Jones DH, Corris S, McDonald S, et al. Poly(dl-lactide-co-glycolide)-encapsulated plasmid DNA elicits systemic and mucosal antibody responses to encoded protein after oral administration. Vaccine. 1997;15:814–817.
  • Zou W, Liu C, Chen Z, et al. Studies on bioadhesive PLGA nanoparticles: a promising gene delivery system for efficient gene therapy to lung cancer. Int J Pharm. 2009;370:187–195.
  • Chumakova OV, Liopo AV, Andreev VG, et al. Composition of PLGA and PEI/DNA nanoparticles improves ultrasound-mediated gene delivery in solid tumors in vivo. Cancer Lett. 2008;261:215–225.
  • Gonzalez H, Hwang SJ, Davis ME. New class of polymers for the delivery of macromolecular therapeutics. Bioconjug Chem. 1999;10:1068–1074.
  • Burckbuchler V, Wintgens V, Leborgne C, et al. Development and characterization of new cyclodextrin polymer-based DNA delivery systems. Bioconjug Chem. 2008;19:2311–2320.
  • Bellocq NC, Pun SH, Jensen GS, et al. Transferrin-containing, cyclodextrin polymer-based particles for tumor-targeted gene delivery. Bioconjug Chem. 2003;14:1122–1132.
  • Huang H, Yu H, Tang G, et al. Low molecular weight polyethylenimine cross-linked by 2-hydroxypropyl-γ-cyclodextrin coupled to peptide targeting HER2 as a gene delivery vector. Biomaterials. 2010;31:1830–1838.
  • Luo D, Saltzman WM. Enhancement of transfection by physical concentration of DNA at the cell surface. Nat Biotech. 2000;18:893–895.
  • Li X, Chen Y, Wang M, et al. A mesoporous silica nanoparticle – PEI – Fusogenic peptide system for siRNA delivery in cancer therapy. Biomaterials. 2013;34:1391–1401.
  • Chen AM, Zhang M, Wei D, et al. Co‐delivery of doxorubicin and Bcl‐2 siRNA by Mesoporous silica nanoparticles enhances the efficacy of chemotherapy in multidrug‐resistant cancer cells. Small. 2009;5:2673–2677.
  • Meng H, Mai WX, Zhang H, et al. Codelivery of an optimal drug/siRNA combination using mesoporous silica nanoparticles to overcome drug resistance in breast cancer in vitro and in vivo. ACS Nano. 2013;7:994–1005.
  • Guerrero-Cázares H, Tzeng SY, Young NP, et al. Biodegradable polymeric nanoparticles show high efficacy and specificity at DNA delivery to human glioblastoma in vitro and in vivo. ACS Nano. 2014;8:5141–5153.
  • Mangraviti A, Tzeng SY, Kozielski KL, et al. Polymeric nanoparticles for Nonviral gene therapy extend brain tumor survival in vivo. ACS Nano. 2015;9:1236–1249.
  • Kozielski KL, Tzeng SY, Mendoza BAHD, et al. Bioreducible cationic polymer-based nanoparticles for efficient and environmentally triggered cytoplasmic siRNA delivery to primary human brain cancer cells. ACS Nano. 2014;8:3232–3241.
  • Gu J, Wang X, Jiang X, et al. Self-assembled carboxymethyl poly (l-histidine) coated poly (β-amino ester)/DNA complexes for gene transfection. Biomaterials. 2012;33:644–658.
  • Kamat CD, Shmueli RB, Connis N, et al. Poly(β-amino ester) nanoparticle delivery of TP53 has activity against small cell lung cancer in vitro and in vivo. Mol Cancer Ther. 2013;12:405–415.
  • Liu Y, Wang Y, Zhang C, et al. Core–shell nanoparticles based on pullulan and poly(β-amino) ester for hepatoma-targeted codelivery of gene and chemotherapy agent. ACS Appl Mater Interfaces. 2014;6:18712–18720.
  • Anderson DG, Peng W, Akinc A, et al. A polymer library approach to suicide gene therapy for cancer. Proc Natl Acad Sci U S A. 2004;101:16028–16033.
  • Dalby B, Cates S, Harris A, et al. Advanced transfection with Lipofectamine 2000 reagent: primary neurons, siRNA, and high-throughput applications. Methods. 2004;33:95–103.
  • Ma Z, Li J, He FT, et al. Cationic lipids enhance siRNA-mediated interferon response in mice. Biochem Biophys Res Commun. 2005;330:755–759.
  • Palliser D, Chowdhury D, Wang QY, et al. An siRNA-based microbicide protects mice from lethal herpes simplex virus 2 infection. Nature. 2006;439:89–94.
  • Smisterová J, Wagenaar A, Stuart MCA, et al. Molecular shape of the cationic lipid controls the structure of cationic lipid/dioleylphosphatidylethanolamine-DNA complexes and the efficiency of gene delivery. J Biol Chem. 2001;276:47615–47622.
  • Koynova R, Wang L, Tarahovsky Y, et al. Lipid phase control of DNA delivery. Bioconjug Chem. 2005;16:1335–1339.
  • Crook K, Stevenson BJ, Dubouchet M, et al. Inclusion of cholesterol in DOTAP transfection complexes increases the delivery of DNA to cells in vitro in the presence of serum. Gene Ther. 1998;5:137–143.
  • Lu JJ, Langer R, Chen JZ. A novel mechanism is involved in cationic lipid-mediated functional siRNA delivery. Mol Pharm. 2009;6:763–771.
  • Umeda M, Nojima S, Inoue K. Effect of lipid composition on HVJ-mediated fusion of glycophorin liposomes to erythrocytes. J Biochem-Tokyo. 1985;97:1301–1310.
  • Hafez IM, Cullis PR. Roles of lipid polymorphism in intracellular delivery. Adv Drug Deliver Rev. 2001;47:139–148.
  • Litzinger DC, Huang L. Phosphatidylethanolamine liposomes - drug delivery, gene-transfer and immunodiagnostic applications. Biochim Biophys Acta. 1992;1113:201–227.
  • Heyes J, Palmer L, Bremner K, et al. Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids. J Control Release. 2005;107:276–287.
  • Moret I, Esteban Peris J, Guillem VM, et al. Stability of PEI–DNA and DOTAP–DNA complexes: effect of alkaline pH, heparin and serum. J Control Release. 2001;76:169–181.
  • Chono S, Li SD, Conwell CC, et al. An efficient and low immunostimulatory nanoparticle formulation for systemic siRNA delivery to the tumor. J Control Release. 2008;131:64–69.
  • Judge AD, Bola G, Lee ACH, et al. Design of noninflammatory synthetic sirna mediating potent gene silencing in vivo. Mol Ther. 2006;13:494–505.
  • Semple SC, Akinc A, Chen J, et al. Rational design of cationic lipids for siRNA delivery. Nat Biotechnol. 2010;28:172–176.
  • Ott G, Singh M, Kazzaz J, et al. A cationic sub-micron emulsion (MF59/DOTAP) is an effective delivery system for DNA vaccines. J Control Release. 2002;79:1–5.
  • Chen C, Okayama H. High-efficiency transformation of mammalian cells by plasmid DNA. Mol Cell Biol. 1987;7:2745–2752.
  • Jordan M, Schallhorn A, Wurm FM. Transfecting mammalian cells: optimization of critical parameters affecting calcium-phosphate precipitate formation. Nucleic Acids Res. 1996;24:596–601.
  • Tolou H. Administration of oligonucleotides to cultured cells by calcium phosphate precipitation method. Anal Biochem. 1993;215:156–158.
  • Sokolova VV, Radtke I, Heumann R, et al. Effective transfection of cells with multi-shell calcium phosphate-DNA nanoparticles. Biomaterials. 2006;27:3147–3153.
  • Kakizawa Y, Furukawa S, Ishii A, et al. Organic-inorganic hybrid-nanocarrier of siRNA constructing through the self-assembly of calcium phosphate and PEG-based block aniomer. J Control Release. 2006;111:368–370.
  • Zhang M, Ishii A, Nishiyama N, et al. PEGylated calcium phosphate nanocomposites as smart environment-sensitive carriers for siRNA delivery. Advanced Mater. 2009;21:3520–3525.
  • Ghosh PS, Kim CK, Han G, et al. Efficient gene delivery vectors by tuning the surface charge density of amino acid-functionalized gold nanoparticles. ACS Nano. 2008;2:2213–2218.
  • Connor EE, Mwamuka J, Gole A, et al. Gold nanoparticles are taken up by human cells but do not cause acute cytotoxicity. Small. 2005;1:325–327.
  • Sandhu KK, McIntosh CM, Simard JM, et al. Gold nanoparticle-mediated transfection of mammalian cells. Bioconjug Chem. 2002;13:3–6.
  • Rosi NL, Giljohann DA, Thaxton CS, et al. Oligonucleotide-modified gold nanoparticles for intracellular gene regulation. Science. 2006;312:1027–1030.
  • Cutler JI, Auyeung E, Mirkin CA. Spherical nucleic acids. J Am Chem Soc. 2012;134:1376–1391.
  • Barnaby SN, Sita TL, Petrosko SH, et al. Therapeutic applications of spherical nucleic acids. In: Mirkin CA, Meade TJ, Petrosko SH, et al., editors. Nanotechnology-based precision tools for the detection and treatment of cancer. Cham: Springer; 2015; 23–50.
  • Han G, You CC, Kim BJ, et al. Light‐regulated release of DNA and its delivery to nuclei by means of photolabile gold nanoparticles. Angewandte Chemie. 2006;118:3237–3241.
  • Elbakry A, Zaky A, Liebl R, et al. Layer-by-layer assembled gold nanoparticles for siRNA delivery. Nano Lett. 2009;9:2059–2064.
  • Lee JS, Green JJ, Love KT, et al. Gold, poly (beta-amino ester) nanoparticles for small interfering RNA delivery. Nano Lett. 2009;9:2402–2406.
  • Derfus AM, Chan WCW, Bhatia SN. Intracellular delivery of quantum dots for live cell labeling and organelle tracking. Advanced Mater. 2004;16:961–966.
  • Gao X, Cui Y, Levenson RM, et al. In vivo cancer targeting and imaging with semiconductor quantum dots. Nat Biotechnol. 2004;22:969–976.
  • Wu X, Liu H, Liu J, et al. Immunofluorescent labeling of cancer marker Her2 and other cellular targets with semiconductor quantum dots. Nat Biotechnol. 2002;21:41–46.
  • Srinivasan C, Lee J, Papadimitrakopoulos F, et al. Labeling and intracellular tracking of functionally active plasmid DNA with semiconductor quantum dots. Mol Ther. 2006;14:192–201.
  • Zhang P, Liu W. ZnO QD@PMAA-co-PDMAEMA nonviral vector for plasmid DNA delivery and bioimaging. Biomaterials. 2010;31:3087–3094.
  • Derfus AM, Chen AA, Min DH, et al. Targeted quantum dot conjugates for siRNA delivery. Bioconjug Chem. 2007;18:1391–1396.
  • Chen AA, Derfus AM, Khetani SR, et al. Quantum dots to monitor RNAi delivery and improve gene silencing. Nucleic Acids Res. 2005;33:e190–e.
  • Slowing II, Vivero-Escoto JL, Wu C-W, et al. Mesoporous silica nanoparticles as controlled release drug delivery and gene transfection carriers. Adv Drug Deliver Rev. 2008;60:1278–1288.
  • Mornet S, Lambert O, Duguet E, et al. The formation of supported lipid bilayers on silica nanoparticles revealed by cryoelectron microscopy. Nano Lett. 2005;5:281–285.
  • Bharali DJ, Klejbor I, Stachowiak EK, et al. Organically modified silica nanoparticles: a nonviral vector for in vivo gene delivery and expression in the brain. Proc Natl Acad Sci U S A. 2005;102:11539–11544.
  • Xia T, Kovochich M, Liong M, et al. Polyethyleneimine coating enhances the cellular uptake of mesoporous silica nanoparticles and allows safe delivery of siRNA and DNA constructs. ACS Nano. 2009;3:3273–3286.
  • Roy I, Ohulchanskyy TY, Bharali DJ, et al. Optical tracking of organically modified silica nanoparticles as DNA carriers: a nonviral, nanomedicine approach for gene delivery. Proc Natl Acad Sci U S A. 2005;102:279–284.
  • Tinsley-Bown AM, Fretwell R, Dowsett AB, et al. Formulation of poly(d,l-lactic-co-glycolic acid) microparticles for rapid plasmid DNA delivery. J Control Release. 2000;66:229–241.
  • Zhou J, Patel TR, Fu M, et al. Octa-functional PLGA nanoparticles for targeted and efficient siRNA delivery to tumors. Biomaterials. 2012;33:583–591.
  • Ziady A-G, Gedeon CR, Miller T, et al. Transfection of airway epithelium by stable PEGylated Poly-L-lysine DNA nanoparticles in vivo. Mol Ther. 2003;8:936–947.
  • Kim J-S, Kim B-I, Maruyama A, et al. A new non-viral DNA delivery vector: the terplex system. J Control Release. 1998;53:175–182.
  • Patil ML, Zhang M, Minko T. Multifunctional triblock nanocarrier (PAMAM-PEG-PLL) for the efficient intracellular siRNA delivery and gene silencing. ACS Nano. 2011;5:1877–1887.
  • Miyata K, Kakizawa Y, Nishiyama N, et al. Block catiomer polyplexes with regulated densities of charge and disulfide cross-linking directed to enhance gene expression. J Am Chem Soc. 2004;126:2355–2361.
  • Christie RJ, Matsumoto Y, Miyata K, et al. Targeted polymeric micelles for siRNA treatment of experimental cancer by intravenous injection. ACS Nano. 2012;6:5174–5189.
  • Godbey WT, Wu KK, Mikos AG. Tracking the intracellular path of poly(ethylenimine)/DNA complexes for gene delivery. Proc Natl Acad Sci. 1999;96:5177–5181.
  • Kunath K, Von Harpe A, Fischer D, et al. Low-molecular-weight polyethylenimine as a non-viral vector for DNA delivery: comparison of physicochemical properties, transfection efficiency and in vivo distribution with high-molecular-weight polyethylenimine. J Control Release. 2003;89:113–125.
  • Kleemann E, Neu M, Jekel N, et al. Nano-carriers for DNA delivery to the lung based upon a TAT-derived peptide covalently coupled to PEG–PEI. J Control Release. 2005;109:299–316.
  • Breunig M, Hozsa C, Lungwitz U, et al. Mechanistic investigation of poly (ethylene imine)-based siRNA delivery: disulfide bonds boost intracellular release of the cargo. J Control Release. 2008;130:57–63.
  • Christensen LV, Chang CW, Kim WJ, et al. Reducible poly(amido ethylenimine)s designed for triggered intracellular gene delivery. Bioconjug Chem. 2006;17:1233–1240.
  • Tang MX, Redemann CT, Szoka FC. In vitro gene delivery by degraded polyamidoamine dendrimers. Bioconjug Chem. 1996;7:703–714.
  • Luo D, Haverstick K, Belcheva N, et al. Poly(ethylene glycol)-conjugated PAMAM dendrimer for biocompatible, high-efficiency DNA delivery. Macromolecules. 2002;35:3456–3462.
  • Jeong JH, Christensen LV, Yockman JW, et al. Reducible poly(amido ethylenimine) directed to enhance RNA interference. Biomaterials. 2007;28:1912–1917.
  • Vader P, Van Der Aa LJ, Engbersen JFJ, et al. Disulfide-based poly(amido amine)s for siRNA delivery: effects of structure on siRNA complexation, cellular uptake, gene silencing and toxicity. Pharm Res. 2011;28:1013–1022.
  • Van Der Aa LJ, Vader P, Storm G, et al. Optimization of poly(amido amine)s as vectors for siRNA delivery. J Control Release. 2011;150:177–186.
  • Vader P, Van Der Aa LJ, Engbersen JFJ, et al. Physicochemical and biological evaluation of siRNA polyplexes based on PEGylated poly (amido amine) s. Pharm Res. 2012;29:352–361.
  • Li S, Purdy WC. Cyclodextrins and their applications in analytical chemistry. Chem Rev. 1992;92:1457–1470.
  • Yang C, Li H, Goh SH, et al. Cationic star polymers consisting of α-cyclodextrin core and oligoethylenimine arms as nonviral gene delivery vectors. Biomaterials. 2007;28:3245–3254.
  • Arima H, Kihara F, Hirayama F, et al. Enhancement of gene expression by polyamidoamine dendrimer conjugates with α-, β-, and γ-cyclodextrins. Bioconjug Chem. 2001;12:476–484.
  • Davis ME. The first targeted delivery of siRNA in humans via a nanoparticle: from concept to clinic. Mol Pharm. 2009;6:659–668.
  • Davis ME, Zuckerman JE, Choi CHJ, et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature. 2010;464:1067–1070.
  • Zuckerman JE, Gritli I, Tolcher A, et al. Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA. Proc Natl Acad Sci. 2014;111:11449–11454.
  • Dufès C, Uchegbu IF, Schätzlein AG. Dendrimers in gene delivery. Adv Drug Deliver Rev. 2005;57:2177–2202.
  • Zinselmeyer BH, Mackay SP, Schatzlein AG, et al. The lower-generation polypropylenimine dendrimers are effective gene-transfer agents. Pharm Res. 2002;19:960–967.
  • Kim T-I, Baek J-U, Zhe Bai C, et al. Arginine-conjugated polypropylenimine dendrimer as a non-toxic and efficient gene delivery carrier. Biomaterials. 2007;28:2061–2067.
  • Taratula O, Garbuzenko OB, Kirkpatrick P, et al. Surface-engineered targeted PPI dendrimer for efficient intracellular and intratumoral siRNA delivery. J Control Release. 2009;140:284–293.
  • Tzeng SY, Higgins LJ, Pomper MG, et al. Student award winner in the Ph.D. category for the 2013 society for biomaterials annual meeting and exposition, april 10-13, 2013, Boston, Massachusetts: biomaterial-mediated cancer-specific DNA delivery to liver cell cultures using synthetic poly(beta-amino ester)s. J Biomed Mater Res A. 2013;101:1837–1845.
  • Shmueli RB, Sunshine JC, Xu Z, et al. Gene delivery nanoparticles specific for human microvasculature and macrovasculature. Nanomedicine. 2012;8:1200–1207.
  • Tzeng SY, Guerrero-Cázares H, Martinez EE, et al. Non-viral gene delivery nanoparticles based on poly(β-amino esters) for treatment of glioblastoma. Biomaterials. 2011;32:5402–5410.
  • Deng X, Zheng N, Song Z, et al. Trigger-responsive, fast-degradable poly(β-amino ester)s for enhanced DNA unpackaging and reduced toxicity. Biomaterials. 2014;35:5006–5015.
  • Hong CA, Eltoukhy AA, Lee H, et al. Dendrimeric siRNA for efficient gene silencing. Angewandte Chemie. 2015;127:6844–6848.
  • Tzeng SY, Green JJ. Subtle changes to polymer structure and degradation mechanism enable highly effective nanoparticles for siRNA and DNA delivery to human brain cancer. Adv Healthc Mater. 2013;2:468–480.
  • Tzeng SY, Hung BP, Grayson WL, et al. Cystamine-terminated poly(beta-amino ester)s for siRNA delivery to human mesenchymal stem cells and enhancement of osteogenic differentiation. Biomaterials. 2012;33:8142–8151.
  • Kozielski KL, Tzeng SY, Green JJ. A bioreducible linear poly(beta-amino ester) for siRNA delivery. Chem Commun. 2013;49:5319–5321.
  • Chen J, Qiu X, Ouyang J, et al. pH and reduction dual-sensitive copolymeric micelles for intracellular doxorubicin delivery. Biomacromolecules. 2011;12:3601–3611.
  • Segovia N, Pont M, Oliva N, et al. Hydrogel doped with nanoparticles for local sustained release of siRNA in breast cancer. Adv Healthc Mater. 2015;4:271–280.
  • Chaichana KL, Zadnik P, Weingart JD, et al. Multiple resections for patients with glioblastoma: prolonging survival. J Neurosurg. 2013;118:812–820.
  • McGirt MJ, Chaichana KL, Gathinji M, et al. Independent association of extent of resection with survival in patients with malignant brain astrocytoma. J Neurosurg. 2009;110:156–162.
  • Stupp R, Mason WP, Van Den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant Temozolomide for glioblastoma. New England J Med. 2005;352:987–996.
  • Cutlar L, Zhou D, Gao Y, et al. Highly branched poly (β-Amino Esters): synthesis and application in gene delivery. Biomacromolecules. 2015;16:2609–2617.
  • Zhou K, Nguyen LH, Miller JB, et al. Modular degradable dendrimers enable small RNAs to extend survival in an aggressive liver cancer model. Proc Natl Acad Sci. 2016;113:520–525.
  • Bhise NS, Shmueli RB, Gonzalez J, et al. A novel assay for quantifying the number of plasmids encapsulated by polymer nanoparticles. Small. 2012;8:367–373.
  • Li S-D, Huang L. Targeted delivery of antisense oligodeoxynucleotide and small interference RNA into lung cancer cells. Mol Pharm. 2006;3:579–588.
  • Chang Kang H, Bae YH. Co-delivery of small interfering RNA and plasmid DNA using a polymeric vector incorporating endosomolytic oligomeric sulfonamide. Biomaterials. 2011;32:4914–4924.
  • Chen Y, Zhu X, Zhang X, et al. Nanoparticles modified with tumor-targeting scFv deliver siRNA and miRNA for cancer therapy. Mol Ther. 2010;18:1650–1656.
  • Bishop CJ, Tzeng SY, Green JJ. Degradable polymer-coated gold nanoparticles for co-delivery of DNA and siRNA. Acta Biomater. 2015;11:393–403.
  • Akinc A, Thomas M, Klibanov AM, et al. Exploring polyethylenimine‐mediated DNA transfection and the proton sponge hypothesis. J Gene Med. 2005;7:657–663.
  • Kichler A, Chillon M, Leborgne C, et al. Intranasal gene delivery with a polyethylenimine–PEG conjugate. J Control Release. 2002;81:379–388.
  • Mao S, Neu M, Germershaus O, et al. Influence of polyethylene glycol chain length on the physicochemical and biological properties of poly(ethylene imine)-graft-poly(ethylene glycol) block copolymer/SiRNA Polyplexes. Bioconjug Chem. 2006;17:1209–1218.
  • Xie J, Chen K, Lee H-Y, et al. Ultrasmall c(RGDyK)-coated Fe3O4 nanoparticles and their specific targeting to integrin αvβ3-rich tumor cells. J Am Chem Soc. 2008;130:7542–7543.
  • Simberg D, Duza T, Park JH, et al. Biomimetic amplification of nanoparticle homing to tumors. Proc Natl Acad Sci. 2007;104:932–936.
  • Harris TJ, Green JJ, Fung PW, et al. Tissue-specific gene delivery via nanoparticle coating. Biomaterials. 2010;31:998–1006.
  • Deltcheva E, Chylinski K, Sharma CM, et al. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature. 2011;471:602–607.
  • Jinek M, Chylinski K, Fonfara I, et al. A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816–821.
  • Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science. 2013;339:823–826.
  • Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/cas systems. Science. 2013;339:819–823.
  • Jinek M, East A, Cheng A, et al. RNA-programmed genome editing in human cells. Elife. 2013;2:e00471.
  • Sun W, Ji W, Hall JM, et al. Self-assembled DNA nanoclews for the efficient delivery of CRISPR–Cas9 for genome editing. Angewandte Chemie. 2015;127:12197–12201.
  • Wiley SR, Schooley K, Smolak PJ, et al. Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity. 1995;3:673–682.
  • Pan G, O’Rourke K, Chinnaiyan AM, et al. The receptor for the cytotoxic ligand TRAIL. Science. 1997;276:111–113.
  • Ashkenazi A, Pai RC, Fong S, et al. Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest. 1999;104:155–162.
  • Kagawa S, He C, Gu J, et al. Antitumor activity and bystander effects of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene. Cancer Res. 2001;61:3330–3338.
  • Ashkenazi A, Dixit VM. Apoptosis control by death and decoy receptors. Curr Opin Cell Biol. 1999;11:255–260.
  • Sanlioglu AD, Dirice E, Aydin C, et al. Surface TRAIL decoy receptor-4 expression is correlated with TRAIL resistance in MCF7 breast cancer cells. BMC Cancer. 2005;5:1–17.
  • Sanlioglu AD, Karacay B, Koksal IT, et al. DcR2 (TRAIL-R4) siRNA and adenovirus delivery of TRAIL (Ad5hTRAIL) break down in vitro tumorigenic potential of prostate carcinoma cells. Cancer Gene Ther. 2007;14:976–984.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.