693
Views
10
CrossRef citations to date
0
Altmetric
Review Articles

Lipoplex-based therapeutics for effective oligonucleotide delivery: a compendious review

, , &
Pages 313-335 | Received 28 Feb 2019, Accepted 31 Jul 2019, Published online: 19 Aug 2019

References

  • Abd Elwakil, M.M., et al., 2019. Lung-endothelium-targeted nanoparticles based on a pH-sensitive lipid and the GALA peptide enable robust gene silencing and the regression of metastatic lung cancer. Advanced functional materials, 29 (18), 1807677.
  • Adams, D.O. and Hamilton, T.A., 1984. The cell biology of macrophage activation. Annual review of immunology, 2, 283–318.
  • Akita, H., et al., 2013. Particle tracking analysis for the intracellular trafficking of nanoparticles modified with African swine fever virus protein p54-derived peptide. Molecular therapy, 21 (2), 309–317.
  • Allison, S.D., Molina, M.C., and Anchordoquy, T.J., 2000. Stabilization of lipid/DNA complexes during the freezing step of the lyophilization process: the particle isolation hypothesis. Biochimica et biophysica acta (Bba) – biomembranes, 1468 (1–2), 127–138.
  • Amadio, M., et al., 2016. Nanosystems based on siRNA silencing HuR expression counteract diabetic retinopathy in rat. Pharmacological research, 111, 713–720.
  • Anchordoquy, T.J., Carpenter, J.F., and Kroll, D.J., 1997. Maintenance of transfection rates and physical characterization of lipid/DNA complexes after freeze-drying and rehydration. Archives of biochemistry and biophysics, 348 (1), 199–206.
  • Anchordoquy, T.J., et al., 2001. Physical stabilization of DNA-based therapeutics. Drug discovery today, 6 (9), 463–470.
  • Andreakos, E., et al., 2009. Amphoteric liposomes enable systemic antigen‐presenting cell-directed delivery of CD40 antisense and are therapeutically effective in experimental arthritis. Arthritis & rheumatism, 60 (4), 994–1005.
  • Anon, 2018. Global Gene Therapy Market to Witness a CAGR of 14.6% during the 2018-2024: Energias Market Research Pvt. Ltd. [online]. Energias Market Research. Available from: https://globenewswire.com/news-release/2018/04/23/1485153/0/en/Global-Gene-Therapy-Market-to-Witness-a-CAGR-of-14-6-during-the-2018-2024-Energias-Market-Research-Pvt-Ltd.html [Accessed 08 February 2018].
  • Anon, 2019. Macugen (pegaptanib sodium injection) [online]. Bausch & Lomb Inc. Available from: http://www.bausch.com/ecp/our-products/rx-pharmaceuticals/rx-pharmaceuticals/macugen-pegaptanib-sodium-injection [Accessed 13 February 2019].
  • Balbino, T.A., et al., 2016. Microfluidic assembly of pDNA/cationic liposome lipoplexes with high pDNA loading for gene delivery. Langmuir, 32 (7), 1799–1807.
  • Belliveau, N.M., et al., 2012. Microfluidic synthesis of highly potent limit-size lipid nanoparticles for in vivo delivery of siRNA. Molecular therapy – nucleic acids, 1, e37.
  • Bloomfield, V.A., 1997. DNA condensation by multivalent cations. Biopolymers, 44 (3), 269–282.
  • Boussif, O., et al., 2001. Enhanced in vitro and in vivo cationic lipid-mediated gene delivery with a fluorinated glycerophosphoethanolamine helper lipid. The journal of gene medicine, 3 (2), 109–114.
  • Brown, D.A., and London, E., 1998. Structure and origin of ordered lipid domains in biological membranes. The journal of membrane biology, 164 (2), 103–114.
  • Burnett, J.C., and Rossi, J.J., 2012. RNA-based therapeutics: current progress and future prospects. Chemistry & biology, 19 (1), 60–71.
  • Burnett, J.C., Rossi, J.J., and Tiemann, K., 2011. Current progress of siRNA/shRNA therapeutics in clinical trials. Biotechnology journal, 6 (9), 1130–1146.
  • Carrière, M., et al., 2002. Optimization of cationic lipid mediated gene transfer: structure-function, physico-chemical, and cellular studies. Journal of liposome research, 12 (1–2), 95–106.
  • Cella, G., et al., 2001. Tissue factor pathway inhibitor release induced by defibrotide and heparins. Clinical and applied thrombosis/hemostasis, 7 (3), 225–228.
  • Chonn, A., Cullis, P.R., and Devine, D.V., 1991. The role of surface charge in the activation of the classical and alternative pathways of complement by liposomes. Journal of immunology, 146 (12), 4234–4241.
  • Clement, J., et al., 2005. Large-scale production of lipoplexes with long shelf-life. European journal of pharmaceutics and biopharmaceutics, 59 (1), 35–43.
  • Coccheri, S., et al., 1988. Acute effects of defibrotide, an experimental antithrombotic agent, on fibrinolysis and blood prostanoids in man. European journal of clinical pharmacology, 35 (2), 151–156.
  • Conner, S.D., and Schmid, S.L., 2003. Regulated portals of entry into the cell. Nature, 422 (6927), 37.
  • Dalby, B., et al., 2004. Advanced transfection with Lipofectamine 2000 reagent: primary neurons, siRNA, and high-throughput applications. Methods, 33 (2), 95–103.
  • Dass, C.R., Walker, T.L., and Burton, M.A., 2002. Liposomes containing cationic dimethyl dioctadecyl ammonium bromide: formulation, quality control, and lipofection efficiency. Drug delivery, 9 (1), 11–18.
  • De Lima, M., et al., 2003. Cationic liposomes for gene delivery: from biophysics to biological applications. Current medicinal chemistry, 10 (14), 1221–1231.
  • Desmet, E., et al., 2016. An elastic liposomal formulation for RNAi-based topical treatment of skin disorders: proof-of-concept in the treatment of psoriasis. International journal of pharmaceutics, 500 (1–2), 268–274.
  • Djuric, Z., et al., 2001. Comparison of iron-catalyzed DNA and lipid oxidation. Journal of biochemical and molecular toxicology, 15 (2), 114–119.
  • Escriou, V., et al., 1998. Cationic lipid-mediated gene transfer: effect of serum on cellular uptake and intracellular fate of lipopolyamine/DNA complexes. Biochimica et biophysica acta (Bba) – biomembranes, 1368 (2), 276–288.
  • Ewert, K., et al., 2005. Cationic lipid–DNA complexes for non-viral gene therapy: relating supramolecular structures to cellular pathways. Expert opinion on biological therapy, 5 (1), 33–53.
  • Ewert, K.K., et al., 2006. A columnar phase of dendritic lipid-based cationic liposome-DNA complexes for gene delivery: hexagonally ordered cylindrical micelles embedded in a DNA honeycomb lattice. Journal of the American chemical society, 128 (12), 3998–4006.
  • Farago, O., Grønbech-Jensen, N., and Pincus, P., 2006. Mesoscale computer modeling of lipid-DNA complexes for gene therapy. Physical review letters, 96 (1), 018102.
  • Farhood, H., Serbina, N., and Huang, L., 1995. The role of dioleoyl phosphatidylethanolamine in cationic liposome mediated gene transfer. Biochimica et biophysica acta (Bba) – biomembranes, 1235 (2), 289–295.
  • Feingold, K.R., 2010. Does inhibition of apolipoprotein B synthesis produce foie gras? Journal of lipid research, 51 (5), 877–878.
  • Felgner, P.L., and Ringold, G., 1989. Cationic liposome-mediated transfection. Nature, 337 (6205), 387.
  • Felgner, P.L., et al., 1997. Nomenclature for synthetic gene delivery systems. Human gene therapy, 8, 511–512.
  • Ferencik, M., Lacko, I., and Devinsky, F., 1990. Immunomodulatory activity of some amphiphilic compounds. J die pharmazie, 45, 695–696.
  • Feuerstein, A., 2016. Sarepta, FDA compromise on plan to secure Duchenne drug approval [online]. https://www.thestreet.com/story/13598536/1/Exondys51-fda-compromise-on-plan-to-secure-duchenne-drug-approval.html [Accessed 15 February 2018].
  • Filion, M.C., and Phillips, N.C., 1997. Anti-inflammatory activity of cationic lipids. British journal of pharmacology, 122 (3), 551–557.
  • Finocchiaro, L.M., et al., 2011. Cytokine-enhanced vaccine and interferon-β plus suicide gene as combined therapy for spontaneous canine sarcomas. Research in veterinary science, 91 (2), 230–234.
  • Foged, C., Nielsen, H.M., and Frokjaer, S., 2007. Liposomes for phospholipase A2 triggered siRNA release: preparation and in vitro test. International journal of pharmaceutics, 331 (2), 160–166.
  • Fortunati, E., et al., 1996. In vitro and in vivo gene transfer to pulmonary cells mediated by cationic liposomes. Biochimica et biophysica acta (Bba) – gene structure and expression, 1306 (1), 55–62.
  • Fynan, E.F., et al., 1993. DNA vaccines: protective immunizations by parenteral, mucosal, and gene-gun inoculations. Proceedings of the national academy of sciences USA, 90 (24), 11478–11482.
  • Gaucheron, J., et al., 2002. Synthesis and properties of novel tetraalkyl cationic lipids. Bioconjugate chemistry, 13 (3), 671–675.
  • Gershon, H., et al., 1993. Mode of formation and structural features of DNA-cationic liposome complexes used for transfection. Biochemistry, 32 (28), 7143–7151.
  • Geusens, B., et al., 2009. Ultradeformable cationic liposomes for delivery of small interfering RNA (siRNA) into human primary melanocytes. Journal of controlled release, 133 (3), 214–220.
  • Geusens, B., et al., 2010. Flexible nanosomes (SECosomes) enable efficient siRNA delivery in cultured primary skin cells and in the viable epidermis of ex vivo human skin. Advanced functional materials, 20 (23), 4077–4090.
  • Gilot, D., et al., 2002. Cationic lipids derived from glycine betaine promote efficient and non-toxic gene transfection in cultured hepatocytes. The journal of gene medicine, 4 (4), 415–427.
  • Ginn, S.L., et al., 2018. Gene therapy clinical trials worldwide to 2017: an update. The journal of gene medicine, 20 (5), e3015.
  • Gonçalves, E., Debs, R.J., and Heath, T.D., 2004. The effect of liposome size on the final lipid/DNA ratio of cationic lipoplexes. Biophysical journal., 86 (3), 1554–1563.
  • Gosangi, M., et al., 2017. Evolution of new “bolaliposomes” using novel α-tocopheryl succinate based cationic lipid and 1, 12-disubstituted dodecane-based bolaamphiphile for efficient gene delivery. Bioconjugate chemistry, 28 (7), 1965–1977.
  • Grabbe, S., et al., 2016. Translating nanoparticulate-personalized cancer vaccines into clinical applications: case study with RNA-lipoplexes for the treatment of melanoma. Nanomedicine, 11 (20), 2723–2734.
  • Group, V.S., 2002. A randomized controlled clinical trial of intravitreous fomivirsen for treatment of newly diagnosed peripheral cytomegalovirus retinitis in patients with aids. American journal of ophthalmology, 133, 467–474.
  • Guo, W., and Lee, R.J., 2000. Efficient gene delivery using anionic liposome-complexed polyplexes (LPDII). Bioscience reports, 20 (5), 419–432.
  • Gustafsson, J., et al., 1995. Complexes between cationic liposomes and DNA visualized by cryo-TEM. Journal of biochimica et biophysica acta (Bba) – biomembranes, 1235 (2), 305–312.
  • Haberman, A., 2018. Alnylam’s patisiran, the first ever FDA- and European Commission-approved RNAi therapeutic [online]. Biopharm Consortium, 2018. Available from: https://biopharmconsortium.com/2018/09/05/alnylams-patisiran-the-first-ever-fda-and-european-commission-approved-rnai-therapeutic/ [Accessed 15 February 2018].
  • Hafez, I.M., Ansell, S., and Cullis, P.R., 2000. Tunable pH-sensitive liposomes composed of mixtures of cationic and anionic lipids. Biophysical journal., 79 (3), 1438–1446.
  • Hafez, I.M., Maurer, N., and Cullis, P.R., 2001. On the mechanism whereby cationic lipids promote intracellular delivery of polynucleic acids. Gene therapy, 8 (15), 1188–1196.
  • Hattori, Y., et al., 2017. Effect of chondroitin sulfate on siRNA biodistribution and gene silencing effect in mice after injection of siRNA lipoplexes. Journal of drug delivery science and technology, 41, 401–409.
  • Hayes, M.E., et al., 2006. Genospheres: self-assembling nucleic acid-lipid nanoparticles suitable for targeted gene delivery. Gene therapy, 13 (7), 646–651.
  • Henle, E.S., Luo, Y., and Linn, S., 1996. Fe2+, Fe3+, and oxygen react with DNA-derived radicals formed during iron-mediated Fenton reactions. Biochemistry, 35 (37), 12212–12219.
  • Hopkins, A.L., and Groom, C.R., 2002. The druggable genome. Nature reviews drug discovery, 1 (9), 727–730.
  • Hoy, S.M., 2018. Patisiran: first global approval. Drugs, 78 (15), 1625–1631.
  • Hua, Y., et al., 2010. Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes & development, 24 (15), 1634–1644.
  • Hulst, R., et al., 2004. Sunfish amphiphiles: conceptually new carriers for DNA delivery. European journal of organic chemistry, 2004 (4), 835–849.
  • Imaoka, T., et al., 1998. In vivo gene transfer into the adult mammalian central nervous system by continuous injection of plasmid DNA–cationic liposome complex. Brain research, 780 (1), 119–128.
  • Inouye, S., 1984. Site-specific cleavage of double-strand DNA by hydroperoxide of linoleic acid. FEBS letters, 172 (2), 231–234.
  • Jahnová, E., et al., 1993. Amphiphilic detergents inhibit production of IgG and IgM by human peripheral blood mononuclear cells. Immunology letters, 39 (1), 71–75.
  • Jeffs, L.B., et al., 2005. A scalable, extrusion-free method for efficient liposomal encapsulation of plasmid DNA. Pharmaceutical research, 22 (3), 362–372.
  • Jin, L., et al., 2014. Current progress in gene delivery technology based on chemical methods and nano-carriers. Theranostics, 4 (3), 240–255.
  • Johnsson, M., Wagenaar, A., and Engberts, J.B., 2003. Sugar-based gemini surfactant with a vesicle-to-micelle transition at acidic pH and a reversible vesicle flocculation near neutral pH. Journal of the American chemical society, 125 (3), 757–760.
  • Kaksonen, M., and Roux, A., 2018. Molecular mechanism and physiological functions of clathrin-mediated endocytosis. Nature reviews molecular cell biology, 19 (5), 313.
  • Kapoor, M., and Burgess, D.J., 2012. Efficient and safe delivery of siRNA using anionic lipids: formulation optimization studies. International journal of pharmaceutics, 432 (1–2), 80–90.
  • Kastner, E., et al., 2014. High-throughput manufacturing of size-tuned liposomes by a new microfluidics method using enhanced statistical tools for characterization. International journal of pharmaceutics, 477 (1–2), 361–368.
  • Keefe, A.D., Pai, S., and Ellington, A., 2010. Aptamers as therapeutics. Nature reviews drug discovery, 9 (7), 537.
  • Kennedy, M.T., et al., 2000. Factors governing the assembly of cationic phospholipid-DNA complexes. Biophysical journal., 78 (3), 1620–1633.
  • Kenny, G.D., et al., 2011. Novel multifunctional nanoparticle mediates siRNA tumour delivery, visualisation and therapeutic tumour reduction in vivo. Journal of controlled release, 149 (2), 111–116.
  • Khan, I.U., et al., 2013. Microfluidics: a focus on improved cancer targeted drug delivery systems. Journal of controlled release, 172 (3), 1065–1074.
  • Khvorova, A., and Watts, J.K., 2017. The chemical evolution of oligonucleotide therapies of clinical utility. Nature biotechnology, 35 (3), 238–248.
  • Kikuchi, I., and Carmona-Ribeiro, A., 2000. Interactions between DNA and synthetic cationic liposomes. The journal of physical chemistry B, 104 (13), 2829–2835
  • Kim, A., et al., 2004. In vitro and in vivo transfection efficiency of a novel ultradeformable cationic liposome. Biomaterials, 25 (2), 305–313.
  • Koltover, I., et al., 1998. An inverted hexagonal phase of cationic liposome-DNA complexes related to DNA release and delivery. Science, 281 (5373), 78–81.
  • Kowzun, M. J., et al., 2018., Topical inhibition of PUMA signaling mitigates radiation injury. Wound repair and regeneration, 26 (6), 413–425.
  • Koynova, R., Wang, L., and Macdonald, R.C., 2006. An intracellular lamellar–nonlamellar phase transition rationalizes the superior performance of some cationic lipid transfection agents. Proceedings of the national academy of sciences USA, 103 (39), 14373–14378.
  • Kreiss, P., et al., 1999. Plasmid DNA size does not affect the physicochemical properties of lipoplexes but modulates gene transfer efficiency. Nucleic acids research, 27 (19), 3792–3798.
  • Krishnaswamy, R., Raghunathan, V.A., and Sood, A.K., 2004. Reentrant phase transitions of DNA-surfactant complexes. Physical review E, 69, 031905.
  • Kulkarni, J.A., Cullis, P.R., and van der Meel, R., 2018. Lipid nanoparticles enabling gene therapies: from concepts to clinical utility. Nucleic acid therapeutics, 28 (3), 146–157.
  • Kunz, W., Testard, F., and Zemb, T., 2009. Correspondence between curvature, packing parameter, and hydrophilic–lipophilic deviation scales around the phase-inversion temperature. Langmuir, 25 (1), 112–115. doi:10.1021/la8028879
  • Kwok, A., and Hart, S.L., 2011. Comparative structural and functional studies of nanoparticle formulations for DNA and siRNA delivery. Nanomedicine, 7 (2), 210–219.
  • Lai, E., and Van Zanten, J.H., 2002. Evidence of lipoplex dissociation in liquid formulations. Journal of pharmaceutical sciences, 91 (5), 1225–1232.
  • Lappalainen, K., et al., 1994. Comparison of cell proliferation and toxicity assays using two cationic liposomes. Pharmaceutical research, 11 (8), 1127–1131.
  • Lasic, D., 1997. Recent developments in medical applications of liposomes: sterically stabilized liposomes in cancer therapy and gene delivery in vivo. Journal of controlled release, 48 (2–3), 203–222.
  • Lee, E.R., et al., 1996. Detailed analysis of structures and formulations of cationic lipids for efficient gene transfer to the lung. Human gene therapy, 7 (14), 1701–1717.
  • Lee, L.K., et al., 2003. Biophysical characterization of an integrin-targeted non-viral vector. Medical science monitor, 9 (1), BR54–BR61.
  • Lee, R.J., and Huang, L., 1996. Folate-targeted, anionic liposome-entrapped polylysine-condensed DNA for tumor cell-specific gene transfer. Journal of biological chemistry, 271 (14), 8481–8487.
  • Le Gall, T., et al., 2010. A novel cationic lipophosphoramide with diunsaturated lipid chains: synthesis, physicochemical properties, and transfection activities. Journal of medicinal chemistry, 53 (4), 1496–1508.
  • Li, P., et al., 2011. A novel cationic liposome formulation for efficient gene delivery via a pulmonary route. Nanotechnology, 22 (24), 245104.
  • Lindow, M., and Kauppinen, S., 2012. Discovering the first microRNA-targeted drug. Journal of cell biology, 199 (3), 407.
  • Litzinger, D.C., 1997. Limitations of cationic liposomes for antisense oligonucleotide delivery in vivo. Journal of liposome research, 7 (1), 51–61.
  • Litzinger, D.C., et al., 1996. Fate of cationic liposomes and their complex with oligonucleotide in vivo. Biochimica et biophysica acta, 1281 (2), 139–149.
  • Liu, S., et al., 2013. Advance in herpes simplex viruses for cancer therapy. Science China life sciences, 56 (4), 298–305.
  • Llères, D., et al., 2004. Dependence of the cellular internalization and transfection efficiency on the structure and physicochemical properties of cationic detergent/DNA/liposomes. The journal of gene medicine, 6 (4), 415–428.
  • Lonez, C., et al., 2009. Fusogenic activity of cationic lipids and lipid shape distribution. Cellular and molecular life sciences, 67 (3), 483–494. doi:10.1007/s00018-009-0197-x
  • Lu, J.J., Langer, R., and Chen, J., 2009. A novel mechanism is involved in cationic lipid-mediated functional siRNA delivery. Molecular pharmaceutics, 6 (3), 763–771.
  • Luo, C., et al., 2016. A novel cationic lipid with intrinsic antitumor activity to facilitate gene therapy of TRAIL DNA. Biomaterials, 102, 239–248.
  • Lv, H., et al., 2006. Toxicity of cationic lipids and cationic polymers in gene delivery. Journal of controlled release, 114 (1), 100–109.
  • Ma, B., et al., 2007. Lipoplex morphologies and their influences on transfection efficiency in gene delivery. Journal of controlled release, 123 (3), 184–194.
  • Madry, H., et al., 2001. Efficacy of cationic liposome-mediated gene transfer to mesangial cells in vitro and in vivo. Journal of molecular medicine, 79 (4), 184–189.
  • Mahato, R.I., et al., 1998. Biodistribution and gene expression of lipid/plasmid complexes after systemic administration. Human gene therapy, 9 (14), 2083–2099.
  • Martens, T.F., et al., 2017. Effect of hyaluronic acid-binding to lipoplexes on intravitreal drug delivery for retinal gene therapy. European journal of pharmaceutical sciences, 103, 27–35.
  • Martinez-Outschoorn, U.E., Sotgia, F., and Lisanti, M.P., 2015. Caveolae and signalling in cancer. Nature reviews cancer, 15 (4), 225.
  • Mcmahon, H.T., and Gallop, J.L., 2005. Membrane curvature and mechanisms of dynamic cell membrane remodelling. Nature, 438 (7068), 590–596.
  • Meidan, V.M., et al., 2006. Specific lipoplex-mediated antisense against Bcl-2 in breast cancer cells: a comparison between different formulations. Journal of liposome research, 16 (1), 27–43.
  • Meisel, J.W., and Gokel, G.W., 2016. A simplified direct lipid mixing lipoplex preparation: comparison of liposomal-, dimethylsulfoxide-, and ethanol-based methods. Scientific reports, 6 (1), 27662.
  • Mendell, J.R., et al., 2016. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Annals of neurology, 79 (2), 257–271.
  • Mignet, N., et al., 2008. Anionic pH-sensitive pegylated lipoplexes to deliver DNA to tumors. International journal of pharmaceutics, 361 (1–2), 194–201.
  • Ming, X., Sato, K., and Juliano, R.L., 2011. Unconventional internalization mechanisms underlying functional delivery of antisense oligonucleotides via cationic lipoplexes and polyplexes. Journal of controlled release, 153 (1), 83–92.
  • Mohr, L., et al., 2001. Cationic liposome-mediated gene delivery to the liver and to hepatocellular carcinomas in mice. Human gene therapy, 12 (7), 799–809.
  • Molina, M.D., Allison, S.D., and Anchordoquy, T., 2001. Maintenance of nonviral vector particle size during the freezing step of the lyophilization process is insufficient for preservation of activity: insight from other structural indicators. Journal of pharmaceutical sciences, 90 (10), 1445–1455.
  • Molina, M.D., and Anchordoquy, T.J., 2008. Degradation of lyophilized lipid/DNA complexes during storage: the role of lipid and reactive oxygen species. Biochimica et biophysica acta (Bba) – biomembranes, 1778 (10), 2119–2126.
  • Nance, E., et al., 2014. Brain penetrating nanoparticles improve paclitaxel efficacy in malignant glioma following local administration. ACS nano, 8 (10), 10655–10664.
  • Ni, R., et al., 2016. Virus-inspired nucleic acid delivery system: linking virus and viral mimicry. Advanced drug delivery reviews, 106, 3–26.
  • Noguchi, A., et al., 1998. Membrane fusion plays an important role in gene transfection mediated by cationic liposomes. FEBS letters, 433 (1–2), 169–173.
  • Ogris, M., et al., 2001. DNA/polyethylenimine transfection particles: influence of ligands, polymer size, and PEGylation on internalization and gene expression. AAPS pharmsciTech, 3 (3), 43.
  • Okada, H., et al., 2012. Gene transfer targeting mouse vestibule using adenovirus and adeno-associated virus vectors. Otology & neurotology, 33 (4), 655–659.
  • Patil, S.D., Rhodes, D.G., and Burgess, D.J., 2004. Anionic liposomal delivery system for DNA transfection. The AAPS journal, 6 (4), e29.
  • Payne, C.K., et al., 2007. Internalization and trafficking of cell surface proteoglycans and proteoglycan-binding ligands. Traffic, 8 (4), 389–401.
  • Pescador, R., et al., 2013. Defibrotide: properties and clinical use of an old/new drug. Vascular pharmacology, 59 (1–2), 1–10.
  • Phadke, A.P., et al., 2011. In vivo safety and antitumor efficacy of bifunctional small hairpin RNAs specific for the human Stathmin 1 oncoprotein. DNA and cell biology, 30 (9), 715–726.
  • Pires, V.B., et al., 2017. Short (16-mer) locked nucleic acid splice-switching oligonucleotides restore dystrophin production in Duchenne Muscular Dystrophy myotubes. Plos one, 12 (7), e0181065.
  • Pitard, B., et al., 1997. Virus-sized self-assembling lamellar complexes between plasmid DNA and cationic micelles promote gene transfer. Proceedings of the national academy of sciences USA, 94 (26), 14412–14417.
  • Potter, H., 1988. Electroporation in biology: methods, applications, and instrumentation. Analytical biochemistry, 174 (2), 361–373.
  • Pozharski, E., and Macdonald, R.C., 2002. Thermodynamics of cationic lipid-DNA complex formation as studied by isothermal titration calorimetry. Biophysical journal., 83 (1), 556–565.
  • Pozharski, E., and Macdonald, R.C., 2003. Lipoplex thermodynamics: determination of DNA-cationic lipoid interaction energies. Biophysical journal., 85 (6), 3969–3978.
  • Rafael, D., et al., 2015. Lipoplexes and polyplexes: gene therapy. In: M. Mishra, ed. Encyclopedia of biomedical polymers and polymeric biomaterials. 1st ed. Boca Raton, FL: CRC Press, 4335–4347.
  • Rao, D.D., et al., 2016. Preclinical justification of pbi-shRNA EWS/FLI1 lipoplex (LPX) treatment for Ewing’s sarcoma. Molecular therapy, 24 (8), 1412–1422.
  • Real, G., et al., 2011. Improvement of lentiviral transfer vectors using cis-acting regulatory elements for increased gene expression. Applied microbiology and biotechnology, 91, 1581.
  • Rehman, Z.U., Hoekstra, D., and Zuhorn, I.S., 2013. Mechanism of polyplex-and lipoplex-mediated delivery of nucleic acids: real-time visualization of transient membrane destabilization without endosomal lysis. ACS nano, 7 (5), 3767–3777.
  • Reiser, A., et al., 2019. Correlation of MRNA delivery timing and protein expression in lipid-based transfection. doi: http://dx.doi.org/10.1101/607986 bioRxiv preprint first posted online April 13 2019; Available from: https://www.biorxiv.org/content/10.1101/607986v1
  • Rejman, J., Bragonzi, A., and Conese, M., 2005. Role of clathrin-and caveolae-mediated endocytosis in gene transfer mediated by lipo-and polyplexes. Molecular therapy, 12 (3), 468–474.
  • Rejman, J., et al., 2004. Size-dependent internalization of particles via the pathways of clathrin-and caveolae-mediated endocytosis. Biochemical journal, 377 (1), 159–169.
  • Richardson, P.G., et al., 2016. Phase 3 trial of defibrotide for the treatment of severe veno-occlusive disease and multi-organ failure. Blood, 127 (13), 1656–1665.
  • Rosenberg, S.A., et al., 1990. Gene transfer into humans – immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. New England journal of medicine, 323 (9), 570–578.
  • Ruckman, J., et al., 1998. Fluoropyrimidine RNA-based aptamers to the 165-amino acid form of vascular endothelial growth factor (VEGF165) inhibition of receptor binding and VEGF-induced vascular permeability through interactions requiring the exon 7-encoded domain. Journal of biological chemistry, 273 (32), 20556–20567.
  • Ruggiero, E., and Richter, S.N., 2018. G-quadruplexes and G-quadruplex ligands: targets and tools in antiviral therapy. Nucleic acids research, 46 (7), 3270–3283.
  • Ruponen, M., et al., 2004. Cell-surface glycosaminoglycans inhibit cation-mediated gene transfer. The journal of gene medicine, 6 (4), 405–414.
  • Sakurai, F., et al., 2001. Effects of erythrocytes and serum proteins on lung accumulation of lipoplexes containing cholesterol or DOPE as a helper lipid in the single-pass rat lung perfusion system. European journal of pharmaceutics and biopharmaceutics, 52 (2), 165–172.
  • Sanders, N.N., et al., 2007. Wanted and unwanted properties of surface PEGylated nucleic acid nanoparticles in ocular gene transfer. Journal of controlled release, 122 (3), 226–235.
  • Scarzello, M., et al., 2005. Sunfish cationic amphiphiles: toward an adaptative lipoplex morphology. Journal of the American chemical society, 127 (29), 10420–10429.
  • Schultheis, B., et al., 2014. First-in-human phase I study of the liposomal RNA interference therapeutic Atu027 in patients with advanced solid tumors. Journal of clinical oncology, 32 (36), 4141–4148.
  • Senior, J.H., Trimble, K.R., and Maskiewicz, R., 1991. Interaction of positively-charged liposomes with blood: implications for their application in vivo. Biochimica et biophysica acta (Bba) – biomembranes, 1070 (1), 173–179.
  • Seville, P.C., Kellaway, I.W., and Birchall, J.C., 2002. Preparation of dry powder dispersions for non-viral gene delivery by freeze-drying and spray-drying. The journal of gene medicine, 4 (4), 428–437.
  • Shen, X., and Corey, D.R., 2018. Chemistry, mechanism and clinical status of antisense oligonucleotides and duplex RNAs. Nucleic acids research, 46 (4), 1584–1600.
  • Silva, J.N., et al., 2014. How multi-step versus one-step preparation method affects the physicochemical properties and transfection efficiency of DNA/DODAB: MO lipoplexes. Journal of applied solution chemistry and modeling, 3, 94–105.
  • Simberg, D., et al., 2001. Phase behavior, DNA ordering, and size instability of cationic lipoplexes. Relevance to optimal transfection activity. Journal of biological chemistry, 276 (50), 47453–47459.
  • Simberg, D., et al., 2003. The role of organ vascularization and lipoplex-serum initial contact in intravenous murine lipofection. Journal of biological chemistry, 278 (41), 39858–39865.
  • Stein, C., et al., 2016b. Defibrotide (defitelio): a new addition to the stockpile of food and drug administration-approved oligonucleotide drugs. Mol Ther Nucleic Acids, 5, e346.
  • Stein, C.A., 2016. Eteplirsen approved for Duchenne muscular dystrophy: the FDA faces a difficult choice. Molecular therapy, 24 (11), 1884–1885.
  • Stein, C.A., and Castanotto, D., 2017. FDA-approved oligonucleotide therapies in 2017. Molecular therapy, 25 (5), 1069–1075.
  • Stephenson, M.L., and Zamecnik, P.C., 1978. Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proceedings of the national academy of sciences USA, 75 (1), 285–288.
  • Sternberg, B., et al., 1998. Ultrastructural characterization of cationic liposome-DNA complexes showing enhanced stability in serum and high transfection activity in vivo. Biochimica et biophysica acta (Bba) – biomembranes, 1375 (1–2), 23–35.
  • Sternberg, B., Sorgi, F.L., and Huang, L., 1994. New structures in complex formation between DNA and cationic liposomes visualized by freeze–fracture electron microscopy. FEBS letters, 356 (2–3), 361–366.
  • Suk, J. S., Xu, Q., Kim, N., Hanes, J., & Ensign, L. M., 2016. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Advanced drug delivery reviews, 99, 28–51.
  • Sullenger, B.A., and Nair, S., 2016. From the RNA world to the clinic. Science, 352 (6292), 1417–1420.
  • Suñé-Pou, M., et al., 2018. Cholesteryl oleate-loaded cationic solid lipid nanoparticles as carriers for efficient gene-silencing therapy. International journal of nanomedicine, 13, 3223–3233.
  • Templeton, N.S., et al., 1997. Improved DNA: liposome complexes for increased systemic delivery and gene expression. Nature biotechnology, 15 (7), 647–652.
  • Tuerk, C., and Gold, L., 1990. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science, 249 (4968), 505–510.
  • Turek, J., et al., 2000. Formulations which increase the size of lipoplexes prevent serum-associated inhibition of transfection. The journal of gene medicine, 2 (1), 32–40.
  • Tzannis, S.T., and Prestrelski, S.J., 1999. Activity–stability considerations of trypsinogen during spray drying: effects of sucrose. Journal of pharmaceutical sciences, 88 (3), 351–358.
  • Uyechi, L.S., et al., 2001. Mechanism of lipoplex gene delivery in mouse lung: binding and internalization of fluorescent lipid and DNA components. Gene therapy, 8 (11), 828–836.
  • Wang, Q., and Wu, Y., 2017. MicroRNA delivery by lipoplexes in lung cancer therapy. The world scientific encyclopedia of nanomedicine and bioengineering I world scientific, 261, 293.
  • Wang, T., et al., 2018. Systematic screening of commonly used commercial transfection reagents towards efficient transfection of single-stranded oligonucleotides. Molecules, 23 (10), 2564.
  • Wang, Z., et al., 2017. Preclinical biodistribution and safety evaluation of a pbi-shRNA STMN1 lipoplex after subcutaneous delivery. Toxicological sciences, 155 (2), 400–408.
  • Weber, C., et al., 2012. Toll-like receptor (TLR) 3 immune modulation by unformulated small interfering RNA or DNA and the role of CD14 (in TLR-mediated effects). Immunology, 136 (1), 64–77.
  • Wolff, J.A., et al., 1990. Direct gene transfer into mouse muscle in vivo. Science, 247 (4949), 1465–1468.
  • Woodle, M.C., 1998. Controlling liposome blood clearance by surface-grafted polymers. Advanced drug delivery reviews, 32 (1–2), 139–152.
  • Wright, M., et al., 1998. β-Galactosidase staining following intracoronary infusion of cationic liposomes in the in vivo rabbit heart is produced by microinfarction rather than effective gene transfer: a cautionary tale. Gene therapy, 5 (3), 301.
  • Wu, S.Y., et al., 2011. Vaginal delivery of siRNA using a novel PEGylated lipoplex-entrapped alginate scaffold system. Journal of controlled release, 155 (3), 418–426.
  • Xu, Y., and Szoka, F.C., 1996. Mechanism of DNA release from cationic liposome/DNA complexes used in cell transfection. Biochemistry, 35 (18), 5616–5623.
  • Xu, Y., et al., 1999. Physicochemical characterization and purification of cationic lipoplexes. Biophysical journal., 77 (1), 341–353.
  • Yamamoto, T., et al., 2011. Antisense drug discovery and development. Future medicinal chemistry, 3 (3), 339–365.
  • Yamauchi, J., et al., 2010. Comparison between a multifunctional envelope-type nano device and lipoplex for delivery to the liver. Biological & pharmaceutical bulletin, 33 (5), 926–929.
  • Yu, J., and Anchordoquy, T.J., 2009a. Effects of moisture content on the storage stability of dried lipoplex formulations. Journal of pharmaceutical sciences, 98 (9), 3278–3289.
  • Yu, J., and Anchordoquy, T.J., 2009b. Synergistic effects of surfactants and sugars on lipoplex stability during freeze-drying and rehydration. Journal of pharmaceutical sciences, 98 (9), 3319–3328.
  • Zabner, J., et al., 1995. Cellular and molecular barriers to gene transfer by a cationic lipid. The journal of biological chemistry, 270 (32), 18997–19007.
  • Zhao, M., et al., 2008. Lipofectamine RNAiMAX: an efficient siRNA transfection reagent in human embryonic stem cells. Molecular biotechnology, 40 (1), 19–26.
  • Zuhorn, I.S., et al., 2005. Nonbilayer phase of lipoplex–membrane mixture determines endosomal escape of genetic cargo and transfection efficiency. Molecular therapy, 11 (5), 801–810.

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.