Barriers and recent advances in non-viral vectors targeting the lungs for cystic fibrosis gene therapy

Document Type : Review Paper

Authors

1 Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Khyber Pakhtunkhwa, Pakistan

2 Department of Microbiology, Kohat University of Science and Technology, Khyber Pakhtunkhwa, Pakistan

Abstract

Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in CFTR genes that affect chloride ion channel. The CF is a good nominee for gene therapy as the asymptomatic carriers are phenotypically normal, and the desired cells are accessible for vector delivery. Gene therapy shows promising effects involving the correction of gene or replacement of the mutant gene with the functional one. Accordingly, various viral and non-viral carriers have been investigated. Although viral vectors are efficient, they have some problems, including mutagenesis, host immune response, higher toxicity, and costliness. On the other hand, non-viral vectors have less toxicity and immunogenic response and are easier to prepare. For a successful gene therapy, the cargo must be delivered to the target site. However, various barriers are faced by non-viral vectors, which make the gene delivery to the target site difficult. Extracellular barrier, which is the first barrier, include nucleases, negatively charged serum proteins, blood cells, and activated immune system. Ciliated epithelium, mucus gel, apical surface glycocalyx, and plasma membrane come in the second category of the barriers. Furthermore, the third category, which is related to the intracellular barriers, includes endosome and lysosome, cytoplasmic nucleases, viscous environment of cytoplasm with different proteins, and finally nuclear membrane. Various approaches have been proposed to increase the systematic delivery of vectors and enhance their efficiency. Some of these approaches include surface coating with inert polymers, modification of surface charge with anionic polymers, and enhancement of endocytosis and reduction of toxicity by using polyethylene glycol. This review paper was conduct to highlight the barriers faced by non-viral vectors when carrying a genetic payload to the lungs. This study also involved the investigation of the strategies and different types of modifications targeted toward the improvement of the efficiency of non-viral vectors.

Keywords


1. Riordan JR, Rommens JM, Kerem B, Alon N, Rozmahel R, Grzelczak Z, Zielenski J, Lok S, Plavsic N, Chou JL. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science. 1989; 245(4922): 1066–1073.
2. Wang Y, Wrennall JA, Cai Z, Li H, Sheppard DN. Understanding how cystic fibrosis mutations disrupt CFTR function: From single molecules to animal models. Int J Biochem. 2014; 52: 47–57.
3. McNally P, Greene C. Cystic fibrosis: a model for precision medicine. Expert Review of Precision Medicine and Drug Development. 2018; 3(2): 107-117.
4. Griesenbach U, Alton EW. Moving forward: Cystic fibrosis gene therapy. Hum Mol Genet. 2013; 22: 52–58.
5. Griesenbach U, Geddes DM, Alton EW. Gene therapy for cystic fibrosis: An example for lung gene therapy. Gene Ther. 2004; 11: 43–50.
6. Kichler A, Chillon M, Leborgne C, Danos O, Frisch B. Intranasal gene delivery with a polyethylenimine-PEG conjugate. J Control Release. 2002; 81(3): 379–388.
7. Di Gioia S, Conese M. Polyethylenimine-mediated gene delivery to the lung and therapeutic applications. Drug Des Devel Ther. 2009; 2: 163–188.
8. Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin RJ, Anderson DG. Non-viral vectors for gene-based therapy. Nat. Rev. Genet.. 2014; 15: 541–555.
9. Pack DW, Hoffman AS, Pun S, Stayton PS. Design and development of polymers for gene delivery. Nat Rev Drug Discov. 2005; 4(7): 581–593.
10. Wiencek J, Lo SF. Advances in the Diagnosis and Management of Cystic Fibrosis in the Genomic Era. Clin Chem. 2018; 64(6): 898-908.
11. Rehman ZU, Hoekstra D, Zuhorn IS. Protein kinase A inhibition modulates the intracellular routing of gene delivery vehicles in HeLa cells, leading to productive transfection. J Control Release. 2011; 156(1): 80–88.
12. Villate-Beitia I, Puras G, Soto-Sánchez C, Agirre M, Ojeda E, Zarate J1, Fernández E, Pedraz JL. Non-viral vectors based on magnetoplexes, lipoplexes and polyplexes for VEGF gene delivery into central nervous system cells. Int J Pharm. 2017; 521: 130–140.
13. Ruponen M, Honkakoski P, Rönkkö S, Pelkonen J, Tammi M, Urtti A. Extracellular and intracellular barriers in non-viral gene delivery. J Control Release. 2003; 93(2): 213–217.
14. Jones CH, Chen CK, Ravikrishnan A, Rane S, Pfeifer BA. Overcoming nonviral gene delivery barriers: Perspective and future. Mol Pharm. 2013; 10(11): 4082–4098.
15. Donnelley M, Parsons D. Gene Therapy for Cystic Fibrosis Lung Disease: Overcoming the Barriers to Translation to the Clinic. Front Pharmacol. 2018; 9: 1381-1381.
16. Zhang Y, Satterlee A, Huang L. In vivo gene delivery by nonviral vectors: Overcoming hurdles. Mol Ther. 2012; 20(7): 1298–1304.
17. Houk BE, Hochhaus G, Hughes JA. Kinetic modeling of plasmid DNA degradation in rat plasma. AAPS PharmSci. 1999; 1(3): 4-9.
18. Mumper RJ, Duguid JG, Anwer K, Barron MK, Nitta H, Rolland AP. Polyvinyl derivatives as novel interactive polymers for controlled gene delivery to muscle. Pharm Res. 1996; 13(5): 701–709.
19. Sternberg B, Hong K, Zheng W, Papahadjopoulos D. Ultrastructural characterization of cationic liposome-DNA complexes showing enhanced stability in serum and high transfection activity in vivo. Biochim Biophys Acta. 1998; 1375(1): 23–35.
20. Ikeda Y, Nagasaki Y. Impacts of PEGylation on the gene and oligonucleotide delivery system. J Appl Polym Sci. 2014; 131: 2-9.
21. Ge Z , Chen Q, Osada K, Liu X, Tockary TA, Uchida S, Dirisala A, Ishii T, Nomoto T, Toh K, Matsumoto Y, Oba M, Kano MR, Itaka K, Kataoka K. Targeted gene delivery by polyplex micelles with crowded PEG palisade and cRGD moiety for systemic treatment of pancreatic tumors. Biomaterials. 2014; 35(10): 3416–3426.
22. Choi YH, Liu F, Kim JS, Choi YK, Park JS, Kim SW. Polyethylene glycol-grafted poly-L-lysine as polymeric gene carder. J Control Release.1998; 54(1): 39–48.
23. Wegman F, Oner FC, Dhert WJ, Alblas J. Non-viral gene therapy for bone tissue engineering. Biotechnol Genet Eng Rev. 2013; 29: 206-220.
24. Semple SC, Chonn A, Cullis PR. Interactions of liposomes and lipid-based carrier systems with blood proteins: Relation to clearance behaviour in vivo. Adv Drug Deliv Rev. 1998; 32: 3–17.
25. Sakurai F, Nishioka T, Saito H, Baba T, Okuda A, Matsumoto O, Taga T, Yamashita F, Takakura Y, Hashida M. Interaction between DNA-cationic liposome complexes and erythrocytes is an important factor in systemic gene transfer via the intravenous route in mice: The role of the neutral helper lipid. Gene Ther. 2001; 8(9): 677–686.
26. Semple SC, Akinc A, Chen J, Sandhu AP, Mui BL, Cho CK, Sah DW, Stebbing D, Crosley EJ, Yaworski E, Hafez IM, Dorkin JR, Qin J, Lam K. Rational design of cationic lipids for siRNA delivery. Nat Biotechnol.2010; 28(2): 172–176.
27. Wang G, Davidson BL, Melchert P, Slepushkin VA, van Es HH, Bodner M, Jolly DJ, McCray PB Jr. Influence of cell polarity on retrovirus-mediated gene transfer to differentiated human airway epithelia. J Virol. 1998; 72(12): 9818–9826.
28. Bartman CM, Egelston J, Ren X, Das R, Phiel CJ. A simple and efficient method for transfecting mouse embryonic stem cells using polyethylenimine. Exp Cell Res. 2015; 1330(1). 178-185
29. Chantarasrivong C Chantarasrivong C, Ueki A, Ohyama R, Unga J, Nakamura S, Nakanishi I, Higuchi Y , Kawakami S, Ando H. Synthesis and Functional Characterization of Novel Sialyl LewisX Mimic-Decorated Liposomes for E-selectin-Mediated Targeting to Inflamed Endothelial Cells. Mol Pharm. 2017; 14(5): 1528–1537.
30. Masson C, Garinot M, Mignet N, Wetzer B, Mailhe P, Scherman D, Bessodes M.. pH-Sensitive PEG lipids containing orthoester linkers: New potential tools for nonviral gene delivery. J Control Release. 2004; 99(3): 423–434.
31. Glover DJ, Lipps HJ, Jans DA. Towards safe, non-viral therapeutic gene expression in humans. Nat Rev Genet. 2005; 6(4): 299–310.
32. Zhou H, Liu D, Liang C. Challenges and strategies: The immune responses in gene therapy. Med Res Rev. 2004; 24(6): 748–761.
33. Dow SW, Fradkin LG, Liggitt DH, Willson AP, Heath TD, Potter TA. Lipid-DNA complexes induce potent activation of innate immune responses and antitumor activity when administered intravenously. J Immunol. 1999; 163(3): 1552–1561.
34. Regnström K, Ragnarsson EGE, Köping-Höggård M, Torstensson E, Nyblom H, Artursson P. PEI - A potent, but not harmless, mucosal immuno-stimulator of mixed T-helper cell response and FasL-mediated cell death in mice. Gene Ther. 2003; 10(18): 1575–1583.
35. Zhao H, Hemmi H, Akira S, Cheng SH, Scheule RK, Yew NS. Contribution of Toll-like receptor 9 signaling to the acute inflammatory response to nonviral vectors. Mol Ther. 2004; 9(2): 241–248.
36. Yasuda K, Ogawa Y, Yamane I, Nishikawa M, Takakura Y. Macrophage activation by a DNA / cationic liposome complex requires endosomal acidification and TLR9-dependent and -independent pathways DNA and vertebrate DNA / cationic liposome. J Leukoc Biol. 2005; 77(1): 71–79.
37. Dams ET, Laverman P, Oyen WJ, Storm G, Scherphof GL, van Der Meer JW, Corstens FH, Boerman OC. Accelerated blood clearance and altered biodistribution of repeated injections of sterically stabilized liposomes. J Pharmacol Exp Ther. 2000; 292(3): 1071–1079.
38. Laverman P, Carstens MG, Boerman OC, Dams ET, Oyen WJ, van Rooijen N, Corstens FH, Storm G.. Factors affecting the accelerated blood clearance of polyethylene glycol-liposomes upon repeated injection. J Pharmacol Exp Ther. 2001; 298(2): 607–612.
39. Semple SC, Harasym TO, Clow KA, Ansell SM, Klimuk SK, Hope MJ. Immunogenicity and rapid blood clearance of liposomes containing polyethylene glycol-lipid conjugates and nucleic Acid. J Pharmacol Exp Ther. 2005; 312(3): 1020–1026.
40. Ishida T, Ichihara M, Wang X, Yamamoto K, Kimura J, Majima E, Kiwada H. Injection of PEGylated liposomes in rats elicits PEG-specific IgM, which is responsible for rapid elimination of a second dose of PEGylated liposomes. J Control Release. 2006; 112(1): 15–25.
41. Ishida T, Wang X, Shimizu T, Nawata K, Kiwada H. PEGylated liposomes elicit an anti-PEG IgM response in a T cell-independent manner. J Control Release. 2007; 122(3): 349–355.
42. Wang X, Ishida T, Kiwada H. Anti-PEG IgM elicited by injection of liposomes is involved in the enhanced blood clearance of a subsequent dose of PEGylated liposomes. J Control Release. 2007; 119(2): 236–244.
43. Vercauteren D, Rejman J, Martens TF, Demeester J, De Smedt SC, Braeckmans K. On the cellular processing of non-viral nanomedicine for nucleic acid delivery: Mechanisms and methods. J Control Release. 2012; 161(2): 566–581.
44. Bahrami B, Mohammadnia-Afrouzi M, Bakhshaei P, Yazdani Y, Ghalamfarsa G, Yousefi M, Sadreddini S, Jadidi-Niaragh F, Hojjat-Farsangi M. Folate-conjugated nanoparticles as a potent therapeutic approach in targeted cancer therapy. Tumour Biol. 2015; 36(8): 5727–5742.
45. Tros de Ilarduya C, Düzgüneş N. Delivery of therapeutic nucleic acids via transferrin and transferrin receptors: lipoplexes and other carriers. Expert Opin Drug Deliv. 2013; 10(11): 1583–1591.
46. Ashraf SQ, Nicholls AM, Wilding JL, Ntouroupi TG, Mortensen NJ, Bodmer WF. Direct and immune mediated antibody targeting of ERBB receptors in a colorectal cancer cell-line panel. Proc Natl Acad Sci U S A. 2012; 109(51): 21046–21051.
47. Oakland M, Sinn PL, McCray PB. Advances in cell and gene-based therapies for cystic fibrosis lung disease. Mol Ther. 2012; 20(6): 1108–1115.
48. Cone RA. Barrier properties of mucus. Adv Drug Deliv Rev. 2009; 61(2): 75–85.
49. Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev. 2016; 99: 28–51.
50. Yeates DB, Aspin N, Levison H, Jones M T, Bryan C. Mucociliary tracheal transport rates in man. J Appl Physiol. 1975; 39: 487–495.
51. Livraghi A and Randell SH. Cystic Fibrosis and Other Respiratory Diseases of Impaired Mucus Clearance. Toxicol Pathol. 2007; 35(1): 116–129.
52. Forier K, Messiaen AS, Raemdonck K, Deschout H, Rejman J, De Baets F, Nelis H, De Smedt SC, Demeester J, Coenye T, Braeckmans K.. Transport of nanoparticles in cystic fibrosis sputum and bacterial biofilms by single-particle tracking microscopy. Nanomedicine (Lond). 2013; 8(6): 935–949.
53. Schuster BS, Suk JS, Woodworth GF, Hanes J. Nanoparticle diffusion in Zespiratory mucus from humans without lung disease. Biomaterials. 2013; 34(13): 3439–3446.
54. Mastorakos P, da Silva AL, Chisholm J, Song E, Choi WK, Boyle MP, Morales MM, Hanes J, Suk JS. Highly compacted biodegradable DNA nanoparticles capable of overcoming the mucus barrier for inhaled lung gene therapy. Proc Natl Acad Sci. 2015; 112(28): 8720–8725.
55. Suk JS, Kim AJ, Trehan K, Schneider CS, Cebotaru L, Woodward OM, Boylan NJ, Boyle MP, Lai SK, Guggino WB, Hanes J. Lung gene therapy with highly compacted DNA nanoparticles that overcome the mucus barrier. J Control Release. 2014; 178(1): 8–17.
56. Schuster BS, Kim AJ, Kays JC, Kanzawa MM, Guggino WB, Boyle MP, Rowe SM, Muzyczka N, Suk JS, Hanes J.. Overcoming the cystic fibrosis sputum barrier to leading adeno-associated virus gene therapy vectors. Mol Ther. 2014; 22(8): 1484–1493.
54. Suk JS, Boylan NJ, Trehan K, Tang BC, Schneider CS, Lin JM, Boyle MP, Zeitlin PL, Lai SK, Cooper MJ, Hanes J.. N-acetylcysteine enhances cystic fibrosis sputum penetration and airway gene transfer by highly compacted DNA nanoparticles. Mol Ther. 2011; 19(11): 1981–1989.
57. Button B, Cai LH, Ehre C, Kesimer M, Hill DB, Sheehan JK, Boucher RC, Rubinstein M. A periciliary brush promotes the lung health by separating the mucus layer from airway epithelia. Science. 2012; 337 (6097): 937–941.
58. Stonebraker JR, Wagner D, Lefensty RW, Burns K, Gendler SJ, Bergelson JM, Boucher RC, O’Neal WK, Pickles RJ. Glycocalyx restricts adenoviral vector access to apical receptors expressed on respiratory epithelium in vitro and in vivo: role for tethered mucins as barriers to lumenal infection. J Virol. 2004; 78(24): 13755–13768.
59. Pickles RJ, Fahrner JA, Petrella JM, Boucher RC, Bergelson JM. Retargeting the coxsackievirus and adenovirus receptor to the apical surface of polarized epithelial cells reveals the glycocalyx as a barrier to adenovirus-mediated gene transfer. J Virol. 2000; 74(13): 6050–6057.
60. Nguyen J, Reul R, Betz T, Dayyoub E, Schmehl T, Gessler T, Bakowsky U, Seeger W, Kissel T. Nanocomposites of lung surfactant and biodegradable cationic nanoparticles improve transfection efficiency to lung cells. J Control Release. 2009; 140(1): 47–54.
61. Densmore CL. Advances in noninvasive pulmonary gene therapy. Curr Drug Deliv. 2006; 3(1): 55-63.
62. Haley B, Frenkel E. Nanoparticles for drug delivery in cancer treatment. Urol Oncol. 2008; 26(1): 57–64.
63. Ernst N, Ulrichskötter S, Schmalix WA, Rädler J, Galneder R, Mayer E, Gersting S, Plank C, Reinhardt D, Rosenecker J. Interaction of liposomal and polycationic transfection complexes with pulmonary surfactant. J Gene Med. 1999; 1(5): 331–340.
64. Haslett C. Granulocyte apoptosis and its role in the resolution and control of lung inflammation. Am J Respir Crit Care Med. 1999; 160: 5-11.
65. Eastman SJ, Lukason MJ, Tousignant JD, Murray H, Lane MD, St George JA, Akita GY, Cherry M, Cheng SH, Scheule RK. A Concentrated and Stable Aerosol Formulation of Cationic Lipid:DNA Complexes Giving High-Level Gene Expression in Mouse Lung. Hum Gene Ther. 1997; 8(6): 765–773.
66. McLachlan G, Davidson H, Holder E, Davies LA, Pringle IA, Sumner-Jones SG, Baker A, Tennant P, Gordon C, Vrettou C, Blundell R, Hyndman L, Stevenson B, Wilson A, Doherty A, Shaw DJ, Coles RL, Painter H, Cheng SH, Scheule RK, Davies JC, Innes JA, Hyde SC, Griesenbach U, Alton EW, Boyd AC, Porteous DJ, Gill DR, Collie DD. Pre-clinical evaluation of three non-viral gene transfer agents for cystic fibrosis after aerosol delivery to the ovine lung. Gene Ther. 2011; 18(10): 996–1005.
67. Bechara C, Sagan S. Cell-penetrating peptides: 20 years later, where do we stand?. FEBS Lett. 2013; 587(12): 1693–1702.
68. Gopal V. Bioinspired peptides as versatile nucleic acid delivery platforms. J Control Release. 2013; 167(3): 323–332.
69. Schellinger JG, Pahang JA, Johnson RN, Chu DS, Sellers DL, Maris DO, Convertine AJ, Stayton PS, Horner PJ, Pun SH. Melittin-grafted HPMA-oligolysine based copolymers for gene delivery. Biomaterials. 2013; 34(9): 2318–2326.
70. Parhiz H, Hashemi M, Ramezani M. Non-biological gene carriers designed for overcoming the major extra-and intracellular hurdles in gene delivery, an updated review. Nanomedicine J. 2015; 2(1):1–20.
71. Lee H, Jeong JH, Park TG. A new gene delivery formulation of polyethylenimine/DNA complexes coated with PEG conjugated fusogenic peptide. J Control Release. 2001; 76(2): 183–192.
72. Swanson JA, Baer SC. Phagocytosis by zippers and triggers. Trends Cell Biol. 1995; 5(3): 89–93.
73. Suk JS, Suh J, Choy K, Lai SK, Fu J, Hanes J. Gene delivery to differentiated neurotypic cells with RGD and HIV Tat peptide functionalized polymeric nanoparticles. Biomaterials. 2006; 27(29): 5143–5150.
74. Parmentier J, Becker MM, Heintz U, Fricker G. Stability of liposomes containing bio-enhancers and tetraether lipids in simulated gastro-intestinal fluids. Int J Pharm. 2011; 405(2): 210–217.
75. Schäfer J, Höbel S, Bakowsky U, Aigner A. Liposome-polyethylenimine complexes for enhanced DNA and siRNA delivery. Biomaterials. 2010; 31(26): 6892–6900.
76. Günther M, Lipka J, Malek A, Gutsch D, Kreyling W, Aigner A. Polyethylenimines for RNAi-mediated gene targeting in vivo and siRNA delivery to the lung. Eur J Pharm Biopharm. 2011; 77(3): 438–449.
77. Wang YQ, Wang F, Deng XQ, Sheng J, Chen SY, Su J. Delivery of Therapeutic AGT shRNA by PEG-Bu for Hypertension Therapy. PLoS One. 2013; 8(7): 68651.
78. Wang M, Wu B, Lu P, Tucker JD, Milazi S, Shah SN, Lu QL. Pluronic-PEI copolymers enhance exon-skipping of 2′-O-methyl phosphorothioate oligonucleotide in cell culture and dystrophic mdx mice. Gene Ther. 2014; 21(1): 52–59.
79. Min SH, Lee DC, Lim MJ, Park HS, Kim DM, Cho CW, Yoon DY, Yeom YI. A composite gene delivery system consisting of polyethylenimine and an amphipathic peptide KALA. J Gene Med. 2006; 8(12): 1425–1434.
80. Leopold PL. Endosomal Escape Pathways for Delivery of Biologicals. Lysosomes: Biology, Diseases, and Therapeutics. 2016; 151(3): 383–407.
81. Pezzoli D, Candiani G. Non-viral gene delivery strategies for gene therapy: A ‘ménage à trois’ among nucleic acids, materials, and the biological environment: Stimuli-responsive gene delivery vectors. J Nanoparticle Res. 2013; 15(3): 269-79.
82. Andaloussi S, Lehto T, Mäger I, Rosenthal-Aizman K, Oprea II, Simonson OE, Sork H, Ezzat K, Copolovici DM, Kurrikoff K, Viola JR, Zaghloul EM, Sillard R, Johansson HJ, Said Hassane F, Guterstam P, Suhorutšenko J, Moreno PM, Oskolkov N, Hälldin J, Tedebark U, Metspalu A, Lebleu B, Lehtiö J, Smith CI, Langel U. Design of a peptide-based vector, PepFect6, for efficient delivery of siRNA in cell culture and systemically in vivo. Nucleic Acids Res. 2011; 39(9): 3972–3987.
83. Lechardeur D, Sohn KJ, Haardt M, Joshi PB, Monck M, Graham RW, Beatty B, Squire J, O’Brodovich H, Lukacs GL. Metabolic instability of plasmid DNA in the cytosol: A potential barrier to gene transfer. Gene Ther. 1999; 6 (4): 482–497.
84. Kao HP, Abney JR, Verkman AS. Determinants of the translational mobility of a small solute in cell cytoplasm. J Cell Biol. 1993; 120(1): 175–184.
85. Lukacs GL, Haggie P, Seksek O, Lechardeur D, Freedman N, Verkman AS. Size-dependent DNA mobility in cytoplasm and nucleus. J Biol Chem. 2000; 275(3): 1625–1629.
86. Dauty E, Verkman AS. Actin cytoskeleton as the principal determinant of size-dependent DNA mobility in cytoplasm: A new barrier for non-viral gene delivery. J Biol Chem. 2005; 280(9): 7823–7828.
87. Gao X, Huang L. Cytoplasmic expression of a reporter gene by co-delivery of T7 RNA polymerase and T7 promoter sequence with cationic liposomes. Nucleic Acids Res. 1993; 21(12): 2867–2872.
88. Zabner J, Fasbender AJ, Moninger T, Poellinger KA, Welsh MJ. Cellular and molecular barriers to gene transfer by a cationic lipid. J Biol Chem. 1995; 270(32): 18997–19007.
89. Mesika A, Kiss V, Brumfeld V, Ghosh G, Reich Z. Enhanced intracellular mobility and nuclear accumulation of DNA plasmids associated with a karyophilic protein. Hum Gene Ther. 2005; 16(2): 200–208.
90. Vaughan EE, Dean DA. Intracellular trafficking of plasmids during transfection is mediated by microtubules. Mol Ther. 2006; 13(2): 422–428.
91. Salman H, Abu-Arish A, Oliel S, Loyter A, Klafter J, Granek R, Elbaum M. Nuclear localization signal peptides induce molecular delivery along microtubules. Biophys J. 2005; 89(3): 2134–2145.
92. Badding MA, Vaughan EE, Dean DA. Transcription factor plasmid binding modulates microtubule interactions and intracellular trafficking during gene transfer. Gene Ther. 2012; 19(3): 338–346.
93. Vaughan EE, Geiger RC, Miller AM, Loh-Marley PL, Suzuki T, Miyata N, Dean DA. Microtubule acetylation through HDAC6 inhibition results in increased transfection efficiency. Mol Ther. 2008; 16(11): 1841–1847.
94. Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015; 33(9): 941–951.
95. Saffari M, Tamaddon AM, Shirazi FH, Oghabian MA, Moghimi HR. Improving cellular uptake and in vivo tumor suppression efficacy of liposomal oligonucleotides by urea as a chemical penetration enhancer. J Gene Med. 2013; 15(1): 12–19.
96. Ogris M, Wagner E. Targeting tumors with non-viral gene delivery systems. Drug Discov Today. 2002; 7(8): 479–485.
97. van der Aa MA, Mastrobattista E, Oosting RS, Hennink WE, Koning GA, Crommelin DJ. The nuclear pore complex: the gateway to successful nonviral gene delivery. Pharm Res. 2006; 23(3): 447–459.
98. Hatayama M, Tomizawa T, Sakai-Kato K, Bouvagnet P, Kose S, Imamoto N, Yokoyama S, Utsunomiya-Tate N, Mikoshiba K, Kigawa T, Aruga J. Functional and structural basis of the nuclear localization signal in the ZIC3 zinc finger domain. Hum Mol Genet. 2008; 17(22): 3459–3473.
99. Yi WJ, Yang J, Li C, Wang HY, Liu CW, Tao L, Cheng SX, Zhuo RX, Zhang XZ. Enhanced nuclear import and transfection efficiency of TAT peptide-based gene delivery systems modified by additional nuclear localization signals. Bioconjug Chem. 2012; 23(1): 125–134.
100. Wittayacom K, Uthaipibull C, Kumpornsin K, Tinikul R, Kochakarn T, Songprakhon P, Chookajorn T. A nuclear targeting system in Plasmodium falciparum. Malar J. 2010; 9(1): 126.
101. Alvisi G, Poon IK, Jans DA. Tumor-specific nuclear targeting: Promises for anti-cancer therapy?. Drug Resist Updat. 2006; 9(2): 40–50.