Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Non-viral vectors for gene-based therapy

Key Points

  • Synthetic delivery vectors have the potential to address many of the limitations of viral vectors, particularly with respect to safety.

  • For systemic delivery of DNA, both lipid-based vectors and polymer-based vectors have been intensively investigated in experimental animals and in clinical trials.

  • The potential of mRNA for therapeutic protein expression in vivo has been investigated as an alternative to DNA-based gene therapy owing to its unique advantages. Recent advances in chemical modifications of mRNA reduce stimulation of the immune system and improve stability when it is delivered in vivo.

  • Small interfering RNA (siRNA) has great therapeutic potential, as it can silence nearly any targeted gene after introduction into cells. Lipid- and polymer-based siRNA nanoparticles and conjugate systems enable successful delivery of chemically modified siRNAs in humans.

  • Levels of microRNA (miRNA) can be restored through the introduction of synthetic miRNAs or mimics as miRNA replacement therapy. One miRNA mimic is currently undergoing evaluation in a Phase I clinical trial.

  • Delivery of genome editing systems — including zinc-finger proteins, transcription activator-like effectors and CRISPR–Cas (clustered regularly interspaced short palindromic repeat–CRISPR-associated) systems — facilitates gene editing at desired sites in the genome. Recent proof-of-concept studies in model organisms have shown that this approach may be used to cure genetic diseases, which is in contrast to the temporary expression or random insertion of a DNA fragment in conventional gene therapy.

Abstract

Gene-based therapy is the intentional modulation of gene expression in specific cells to treat pathological conditions. This modulation is accomplished by introducing exogenous nucleic acids such as DNA, mRNA, small interfering RNA (siRNA), microRNA (miRNA) or antisense oligonucleotides. Given the large size and the negative charge of these macromolecules, their delivery is typically mediated by carriers or vectors. In this Review, we introduce the biological barriers to gene delivery in vivo and discuss recent advances in material sciences, nanotechnology and nucleic acid chemistry that have yielded promising non-viral delivery systems, some of which are currently undergoing testing in clinical trials. The diversity of these systems highlights the recent progress of gene-based therapy using non-viral approaches.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Barriers to successful in vivo delivery of nucleic acids using non-viral vectors.
Figure 2: Chemical structures of non-viral DNA vectors.
Figure 3: Structure of non-viral siRNA vectors.
Figure 4: Potential use of non-viral vectors for the delivery of precise genome editing systems.

Similar content being viewed by others

References

  1. Ginn, S. L., Alexander, I. E., Edelstein, M. L., Abedi, M. R. & Wixon, J. Gene therapy clinical trials worldwide to 2012 — an update. J. Gene Med. 15, 65–77 (2013).

    CAS  PubMed  Google Scholar 

  2. Kay, M. A. State-of-the-art gene-based therapies: the road ahead. Nature Rev. Genet. 12, 316–328 (2011).

    CAS  PubMed  Google Scholar 

  3. Mingozzi, F. & High, K. A. Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nature Rev. Genet. 12, 341–355 (2011).

    CAS  PubMed  Google Scholar 

  4. Baum, C., Kustikova, O., Modlich, U., Li, Z. & Fehse, B. Mutagenesis and oncogenesis by chromosomal insertion of gene transfer vectors. Hum. Gene Ther. 17, 253–263 (2006).

    CAS  PubMed  Google Scholar 

  5. Bessis, N., GarciaCozar, F. J. & Boissier, M. C. Immune responses to gene therapy vectors: influence on vector function and effector mechanisms. Gene Ther. 11 (Suppl. 1), S10–S17 (2004).

    CAS  PubMed  Google Scholar 

  6. Waehler, R., Russell, S. J. & Curiel, D. T. Engineering targeted viral vectors for gene therapy. Nature Rev. Genet. 8, 573–587 (2007).

    CAS  PubMed  Google Scholar 

  7. Thomas, C. E., Ehrhardt, A. & Kay, M. A. Progress and problems with the use of viral vectors for gene therapy. Nature Rev. Genet. 4, 346–358 (2003).

    CAS  PubMed  Google Scholar 

  8. Bouard, D., Alazard-Dany, D. & Cosset, F. L. Viral vectors: from virology to transgene expression. Br. J. Pharmacol. 157, 153–165 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Pack, D. W., Hoffman, A. S., Pun, S. & Stayton, P. S. Design and development of polymers for gene delivery. Nature Rev. Drug Discov. 4, 581–593 (2005).

    CAS  Google Scholar 

  10. Mintzer, M. A. & Simanek, E. E. Nonviral vectors for gene delivery. Chem. Rev. 109, 259–302 (2009).

    CAS  PubMed  Google Scholar 

  11. Putnam, D. Polymers for gene delivery across length scales. Nature Mater. 5, 439–451 (2006).

    CAS  Google Scholar 

  12. Davis, M. E. The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Mol. Pharmaceut. 6, 659–668 (2009).

    CAS  Google Scholar 

  13. Gonzalez, H., Hwang, S. J. & Davis, M. E. New class of polymers for the delivery of macromolecular therapeutics. Bioconjug. Chem. 10, 1068–1074 (1999).

    CAS  PubMed  Google Scholar 

  14. Semple, S. C. et al. Rational design of cationic lipids for siRNA delivery. Nature Biotech. 28, 172–176 (2010). This study carries out a rational design of a siRNA delivery vector and achieved significant improvement in delivery efficiency.

    CAS  Google Scholar 

  15. Love, K. T. et al. Lipid-like materials for low-dose, in vivo gene silencing. Proc. Natl Acad. Sci. USA 107, 1864–1869 (2010).

    CAS  PubMed  Google Scholar 

  16. Monopoli, M. P., Aberg, C., Salvati, A. & Dawson, K. A. Biomolecular coronas provide the biological identity of nanosized materials. Nature Nanotechnol. 7, 779–786 (2012).

    CAS  Google Scholar 

  17. Lee, H. et al. Molecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNA delivery. Nature Nanotechnol. 7, 389–393 (2012).

    CAS  Google Scholar 

  18. Kariko, K., Buckstein, M., Ni, H. & Weissman, D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 165–175 (2005).

    CAS  PubMed  Google Scholar 

  19. Anderson, B. R. et al. Nucleoside modifications in RNA limit activation of 2′-5′-oligoadenylate synthetase and increase resistance to cleavage by RNase L. Nucleic Acids Res. 39, 9329–9338 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Kariko, K., Muramatsu, H., Keller, J. M. & Weissman, D. Increased erythropoiesis in mice injected with submicrogram quantities of pseudouridine-containing mRNA encoding erythropoietin. Mol. Ther. 20, 948–953 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Deleavey, G. F., Watts, J. K. & Damha, M. J. Chemical modification of siRNA. Curr. Protoc. Nucleic Acid Chem. 39, 16.3.1–16.3.22 (2009).

    Google Scholar 

  22. Mehier-Humbert, S. & Guy, R. H. Physical methods for gene transfer: improving the kinetics of gene delivery into cells. Adv. Drug Deliv. Rev. 57, 733–753 (2005).

    CAS  PubMed  Google Scholar 

  23. Wells, D. J. Gene therapy progress and prospects: electroporation and other physical methods. Gene Ther. 11, 1363–1369 (2004).

    CAS  PubMed  Google Scholar 

  24. Newman, C. M. & Bettinger, T. Gene therapy progress and prospects: ultrasound for gene transfer. Gene Ther. 14, 465–475 (2007).

    CAS  PubMed  Google Scholar 

  25. Plank, C. et al. The magnetofection method: using magnetic force to enhance gene delivery. Biol. Chem. 384, 737–747 (2003).

    CAS  PubMed  Google Scholar 

  26. Zhang, G., Budker, V. G., Ludtke, J. J. & Wolff, J. A. Naked DNA gene transfer in mammalian cells. Methods Mol. Biol. 245, 251–264 (2004).

    CAS  PubMed  Google Scholar 

  27. Li, W. & Szoka, F. C. Jr. Lipid-based nanoparticles for nucleic acid delivery. Pharm. Res. 24, 438–449 (2007).

    PubMed  Google Scholar 

  28. Thomas, M. & Klibanov, A. M. Non-viral gene therapy: polycation-mediated DNA delivery. Appl. Microbiol. Biotechnol. 62, 27–34 (2003).

    CAS  PubMed  Google Scholar 

  29. Lee, C. C., MacKay, J. A., Frechet, J. M. & Szoka, F. C. Designing dendrimers for biological applications. Nature Biotech. 23, 1517–1526 (2005).

    CAS  Google Scholar 

  30. Discher, D. E. & Ahmed, F. Polymersomes. Annu. Rev. Biomed. Engineer. 8, 323–341 (2006).

    CAS  Google Scholar 

  31. Martin, M. E. & Rice, K. G. Peptide-guided gene delivery. AAPS J. 9, E18–29 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Sokolova, V. & Epple, M. Inorganic nanoparticles as carriers of nucleic acids into cells. Angew. Chem. Int. Ed. Engl. 47, 1382–1395 (2008).

    CAS  PubMed  Google Scholar 

  33. Kawabata, K., Takakura, Y. & Hashida, M. The fate of plasmid DNA after intravenous injection in mice: involvement of scavenger receptors in its hepatic uptake. Pharm. Res. 12, 825–830 (1995).

    CAS  PubMed  Google Scholar 

  34. McManus, J. J., Radler, J. O. & Dawson, K. A. Observation of a rectangular columnar phase in a DNA–calcium–zwitterionic lipid complex. J. Am. Chem. Soc. 126, 15966–15967 (2004).

    CAS  PubMed  Google Scholar 

  35. McManus, J. J., Rädler, J. O. & Dawson, K. A. Does calcium turn a zwitterionic lipid cationic? J. Phys. Chem. B 107, 9869–9875 (2003).

    CAS  Google Scholar 

  36. Koltover, I., Salditt, T., Radler, J. O. & Safinya, C. R. An inverted hexagonal phase of cationic liposome-DNA complexes related to DNA release and delivery. Science 281, 78–81 (1998). This paper elucidates structural considerations involved in the endosomal escape of DNA mediated by liposomal nanoparticles.

    CAS  PubMed  Google Scholar 

  37. Wiethoff, C. M. & Middaugh, C. R. Barriers to nonviral gene delivery. J. Pharm. Sci. 92, 203–217 (2003).

    CAS  PubMed  Google Scholar 

  38. Morille, M., Passirani, C., Vonarbourg, A., Clavreul, A. & Benoit, J. P. Progress in developing cationic vectors for non-viral systemic gene therapy against cancer. Biomaterials 29, 3477–3496 (2008).

    CAS  PubMed  Google Scholar 

  39. Capecchi, M. R. High efficiency transformation by direct microinjection of DNA into cultured mammalian cells. Cell 22, 479–488 (1980).

    CAS  PubMed  Google Scholar 

  40. Miller, A. M. & Dean, D. A. Tissue-specific and transcription factor-mediated nuclear entry of DNA. Adv. Drug Deliv. Rev. 61, 603–613 (2009).

    CAS  PubMed  Google Scholar 

  41. Varga, C. M. et al. Quantitative comparison of polyethylenimine formulations and adenoviral vectors in terms of intracellular gene delivery processes. Gene Ther. 12, 1023–1032 (2005).

    CAS  PubMed  Google Scholar 

  42. Dinh, A. T., Pangarkar, C., Theofanous, T. & Mitragotri, S. Understanding intracellular transport processes pertinent to synthetic gene delivery via stochastic simulations and sensitivity analyses. Biophys. J. 92, 831–846 (2007).

    CAS  PubMed  Google Scholar 

  43. Wasungu, L. & Hoekstra, D. Cationic lipids, lipoplexes and intracellular delivery of genes. J. Control. Release 116, 255–264 (2006).

    CAS  PubMed  Google Scholar 

  44. Godbey, W. T., Wu, K. K. & Mikos, A. G. Tracking the intracellular path of poly(ethylenimine)/DNA complexes for gene delivery. Proc. Natl Acad. Sci. USA 96, 5177–5181 (1999).

    CAS  PubMed  Google Scholar 

  45. Breunig, M. et al. Gene delivery with low molecular weight linear polyethylenimines. J. Gene Med. 7, 1287–1298 (2005).

    CAS  PubMed  Google Scholar 

  46. Schaffer, D. V., Fidelman, N. A., Dan, N. & Lauffenburger, D. A. Vector unpacking as a potential barrier for receptor-mediated polyplex gene delivery. Biotechnol. Bioeng. 67, 598–606 (2000).

    CAS  PubMed  Google Scholar 

  47. Cohen, R. N., van der Aa, M. A., Macaraeg, N., Lee, A. P. & Szoka, F. C. Jr. Quantification of plasmid DNA copies in the nucleus after lipoplex and polyplex transfection. J. Controlled Release 135, 166–174 (2009).

    CAS  Google Scholar 

  48. Gill, D. R., Pringle, I. A. & Hyde, S. C. Progress and prospects: the design and production of plasmid vectors. Gene Ther. 16, 165–171 (2009).

    CAS  PubMed  Google Scholar 

  49. Gill, D. R. et al. Increased persistence of lung gene expression using plasmids containing the ubiquitin C or elongation factor 1α promoter. Gene Ther. 8, 1539–1546 (2001).

    CAS  PubMed  Google Scholar 

  50. Wooddell, C. I., Reppen, T., Wolff, J. A. & Herweijer, H. Sustained liver-specific transgene expression from the albumin promoter in mice following hydrodynamic plasmid DNA delivery. J. Gene Med. 10, 551–563 (2008).

    CAS  PubMed  Google Scholar 

  51. Miao, C. H. et al. Inclusion of the hepatic locus control region, an intron, and untranslated region increases and stabilizes hepatic factor IX gene expression in vivo but not in vitro. Mol. Ther. 1, 522–532 (2000).

    CAS  PubMed  Google Scholar 

  52. Argyros, O. et al. Persistent episomal transgene expression in liver following delivery of a scaffold/matrix attachment region containing non-viral vector. Gene Ther. 15, 1593–1605 (2008).

    CAS  PubMed  Google Scholar 

  53. Kreiss, P. et al. Plasmid DNA size does not affect the physicochemical properties of lipoplexes but modulates gene transfer efficiency. Nucleic Acids Res. 27, 3792–3798 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Darquet, A. M. et al. Minicircle: an improved DNA molecule for in vitro and in vivo gene transfer. Gene Ther. 6, 209–218 (1999).

    CAS  PubMed  Google Scholar 

  55. Kay, M. A., He, C. Y. & Chen, Z. Y. A robust system for production of minicircle DNA vectors. Nature Biotech. 28, 1287–1289 (2010).

    CAS  Google Scholar 

  56. Ehrhardt, A., Xu, H., Huang, Z., Engler, J. A. & Kay, M. A. A direct comparison of two nonviral gene therapy vectors for somatic integration: in vivo evaluation of the bacteriophage integrase phiC31 and the Sleeping Beauty transposase. Mol. Ther. 11, 695–706 (2005).

    CAS  PubMed  Google Scholar 

  57. Wu, S. C. et al. PiggyBac is a flexible and highly active transposon as compared to Sleeping Beauty, Tol2, and Mos1 in mammalian cells. Proc. Natl Acad. Sci. USA 103, 15008–15013 (2006).

    CAS  PubMed  Google Scholar 

  58. Aronovich, E. L., McIvor, R. S. & Hackett, P. B. The Sleeping Beauty transposon system: a non-viral vector for gene therapy. Hum. Mol. Genet. 20, R14–R20 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Fraley, R., Subramani, S., Berg, P. & Papahadjopoulos, D. Introduction of liposome-encapsulated SV40 DNA into cells. J. Biol. Chem. 255, 10431–10435 (1980).

    CAS  PubMed  Google Scholar 

  60. Felgner, P. L. et al. Lipofection: a highly efficient, lipid-mediated DNA-transfection procedure. Proc. Natl Acad. Sci. USA 84, 7413–7417 (1987). References 59 and 60 are the earliest studies to show lipid-mediated DNA delivery in vitro.

    CAS  PubMed  Google Scholar 

  61. Whitehead, K. A., Langer, R. & Anderson, D. G. Knocking down barriers: advances in siRNA delivery. Nature Rev. Drug Discov. 8, 129–138 (2009).

    CAS  Google Scholar 

  62. Lonez, C., Vandenbranden, M. & Ruysschaert, J. M. Cationic liposomal lipids: from gene carriers to cell signaling. Prog. Lipid Res. 47, 340–347 (2008).

    CAS  PubMed  Google Scholar 

  63. Hersey, P. & Gallagher, S. Intralesional immunotherapy for melanoma. J. Surg. Oncol. 109, 320–326 (2014).

    CAS  PubMed  Google Scholar 

  64. Olins, D. E., Olins, A. L. & Von Hippel, P. H. Model nucleoprotein complexes: studies on the interaction of cationic homopolypeptides with DNA. J. Mol. Biol. 24, 157–176 (1967).

    CAS  PubMed  Google Scholar 

  65. Laemmli, U. K. Characterization of DNA condensates induced by poly(ethylene oxide) and polylysine. Proc. Natl Acad. Sci. USA 72, 4288–4292 (1975).

    CAS  PubMed  Google Scholar 

  66. Wu, G. Y. & Wu, C. H. Receptor-mediated in vitro gene transformation by a soluble DNA carrier system. J. Biol. Chem. 262, 4429–4432 (1987).

    CAS  PubMed  Google Scholar 

  67. Wu, G. Y. & Wu, C. H. Receptor-mediated gene delivery and expression in vivo. J. Biol. Chem. 263, 14621–14624 (1988). References 66 and 67 are among the first to investigate the possibility of targeted non-viral nucleic acid delivery in vitro and in vivo.

    CAS  PubMed  Google Scholar 

  68. Choi, Y. H. et al. Polyethylene glycol-grafted poly-L-lysine as polymeric gene carrier. J. Control. Release 54, 39–48 (1998).

    PubMed  Google Scholar 

  69. Kim, S. W. Polylysine copolymers for gene delivery. Cold Spring Harb.Protoc. 2012, 433–438 (2012).

    PubMed  Google Scholar 

  70. Alexis, F., Pridgen, E., Molnar, L. K. & Farokhzad, O. C. Factors affecting the clearance and biodistribution of polymeric nanoparticles. Mol. Pharm. 5, 505–515 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Bazile, D. et al. Stealth Me. PEG–PLA nanoparticles avoid uptake by the mononuclear phagocytes system. J. Pharm. Sci. 84, 493–498 (1995).

    CAS  PubMed  Google Scholar 

  72. Konstan, M. W. et al. Compacted DNA nanoparticles administered to the nasal mucosa of cystic fibrosis subjects are safe and demonstrate partial to complete cystic fibrosis transmembrane regulator reconstitution. Hum. Gene Ther. 15, 1255–1269 (2004).

    CAS  PubMed  Google Scholar 

  73. Lungwitz, U., Breunig, M., Blunk, T. & Gopferich, A. Polyethylenimine-based non-viral gene delivery systems. Eur. J. Pharm. Biopharm. 60, 247–266 (2005).

    CAS  PubMed  Google Scholar 

  74. Boussif, O. et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc. Natl Acad. Sci. USA 92, 7297–7301 (1995). This is the first paper to show that PEI can facilitate DNA transfection in vitro and in vivo.

    CAS  PubMed  Google Scholar 

  75. Godbey, W. T., Wu, K. K. & Mikos, A. G. Size matters: molecular weight affects the efficiency of poly(ethylenimine) as a gene delivery vehicle. J. Biomed. Mater. Res. 45, 268–275 (1999).

    CAS  PubMed  Google Scholar 

  76. Wightman, L. et al. Different behavior of branched and linear polyethylenimine for gene delivery in vitro and in vivo. J. Gene Med. 3, 362–372 (2001).

    CAS  PubMed  Google Scholar 

  77. Goula, D. et al. Polyethylenimine-based intravenous delivery of transgenes to mouse lung. Gene Ther. 5, 1291–1295 (1998).

    CAS  PubMed  Google Scholar 

  78. Kircheis, R., Wightman, L. & Wagner, E. Design and gene delivery activity of modified polyethylenimines. Adv. Drug Deliv. Rev. 53, 341–358 (2001).

    CAS  PubMed  Google Scholar 

  79. Coll, J. L. et al. In vivo delivery to tumors of DNA complexed with linear polyethylenimine. Hum. Gene Ther. 10, 1659–1666 (1999).

    CAS  PubMed  Google Scholar 

  80. Lv, H., Zhang, S., Wang, B., Cui, S. & Yan, J. Toxicity of cationic lipids and cationic polymers in gene delivery. J. Control. Release 114, 100–109 (2006).

    CAS  PubMed  Google Scholar 

  81. Nguyen, H. K. et al. Evaluation of polyether-polyethyleneimine graft copolymers as gene transfer agents. Gene Ther. 7, 126–138 (2000).

    CAS  PubMed  Google Scholar 

  82. Petersen, H. et al. Polyethylenimine-graft-poly(ethylene glycol) copolymers: influence of copolymer block structure on DNA complexation and biological activities as gene delivery system. Bioconjug. Chem. 13, 845–854 (2002).

    CAS  PubMed  Google Scholar 

  83. Breunig, M., Lungwitz, U., Liebl, R. & Goepferich, A. Breaking up the correlation between efficacy and toxicity for nonviral gene delivery. Proc. Natl Acad. Sci. USA 104, 14454–14459 (2007).

    CAS  PubMed  Google Scholar 

  84. Thomas, M. & Klibanov, A. M. Enhancing polyethylenimine's delivery of plasmid DNA into mammalian cells. Proc. Natl Acad. Sci. USA 99, 14640–14645 (2002).

    CAS  PubMed  Google Scholar 

  85. Fortune, J. A., Novobrantseva, T. I. & Klibanov, A. M. Highly effective gene transfection in vivo by alkylated polyethylenimine. J. Drug Deliv. 2011, 204058 (2011).

    PubMed  PubMed Central  Google Scholar 

  86. Zangi, L. et al. Modified mRNA directs the fate of heart progenitor cells and induces vascular regeneration after myocardial infarction. Nature Biotech. 31, 898–907 (2013).

    CAS  Google Scholar 

  87. Kormann, M. S. et al. Expression of therapeutic proteins after delivery of chemically modified mRNA in mice. Nature Biotech. 29, 154–157 (2011). References 86 and 87 show the therapeutic potential of delivery of modified mRNA in vivo.

    CAS  Google Scholar 

  88. Su, X., Fricke, J., Kavanagh, D. G. & Irvine, D. J. In vitro and in vivo mRNA delivery using lipid-enveloped pH-responsive polymer nanoparticles. Mol. Pharm. 8, 774–787 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Phua, K. K., Leong, K. W. & Nair, S. K. Transfection efficiency and transgene expression kinetics of mRNA delivered in naked and nanoparticle format. J. Control. Release 166, 227–233 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Kanasty, R. L., Whitehead, K. A., Vegas, A. J. & Anderson, D. G. Action and reaction: the biological response to siRNA and its delivery vehicles. Mol. Ther. 20, 513–524 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Layzer, J. M. et al. In vivo activity of nuclease-resistant siRNAs. RNA 10, 766–771 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Jackson, A. L. & Linsley, P. S. Recognizing and avoiding siRNA off-target effects for target identification and therapeutic application. Nature Rev. Drug Discov. 9, 57–67 (2010).

    CAS  Google Scholar 

  93. Nguyen, D. N. et al. Lipid-derived nanoparticles for immunostimulatory RNA adjuvant delivery. Proc. Natl Acad. Sci. USA 109, E797–E803 (2012).

    CAS  PubMed  Google Scholar 

  94. Whitehead, K. A., Dahlman, J. E., Langer, R. S. & Anderson, D. G. Silencing or stimulation? siRNA delivery and the immune system. Annu. Rev. Chem. Biomol. Eng. 2, 77–96 (2011).

    CAS  PubMed  Google Scholar 

  95. Wang, A. Z., Langer, R. & Farokhzad, O. C. Nanoparticle delivery of cancer drugs. Annu. Rev. Med. 63, 185–198 (2012).

    CAS  PubMed  Google Scholar 

  96. Wolfrum, C. et al. Mechanisms and optimization of in vivo delivery of lipophilic siRNAs. Nature Biotech. 25, 1149–1157 (2007).

    CAS  Google Scholar 

  97. Akinc, A. et al. Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol. Ther. 18, 1357–1364 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Malek, A. et al. In vivo pharmacokinetics, tissue distribution and underlying mechanisms of various PEI(-PEG)/siRNA complexes. Toxicol. Appl. Pharmacol. 236, 97–108 (2009).

    CAS  PubMed  Google Scholar 

  99. Huang, Y. et al. Elimination pathways of systemically delivered siRNA. Mol. Ther. 19, 381–385 (2011).

    CAS  PubMed  Google Scholar 

  100. Rozema, D. B. et al. Dynamic PolyConjugates for targeted in vivo delivery of siRNA to hepatocytes. Proc. Natl Acad. Sci. USA 104, 12982–12987 (2007).

    CAS  PubMed  Google Scholar 

  101. Kanasty, R., Dorkin, J. R., Vegas, A. & Anderson, D. Delivery materials for siRNA therapeutics. Nature Mater. 12, 967–977 (2013).

    CAS  Google Scholar 

  102. Zuckerman, J. E., Choi, C. H., Han, H. & Davis, M. E. Polycation–siRNA nanoparticles can disassemble at the kidney glomerular basement membrane. Proc. Natl Acad. Sci. USA 109, 3137–3142 (2012).

    CAS  PubMed  Google Scholar 

  103. Naeye, B. et al. In vivo disassembly of IV administered siRNA matrix nanoparticles at the renal filtration barrier. Biomaterials 34, 2350–2358 (2013).

    CAS  PubMed  Google Scholar 

  104. Aird, W. C. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circul. Res. 100, 158–173 (2007).

    CAS  Google Scholar 

  105. Wisse, E., Jacobs, F., Topal, B., Frederik, P. & De Geest, B. The size of endothelial fenestrae in human liver sinusoids: implications for hepatocyte-directed gene transfer. Gene Ther. 15, 1193–1199 (2008).

    CAS  PubMed  Google Scholar 

  106. Maeda, H. Tumor-selective delivery of macromolecular drugs via the EPR effect: background and future prospects. Bioconjug. Chem. 21, 797–802 (2010).

    CAS  PubMed  Google Scholar 

  107. Yu, B., Zhao, X., Lee, L. J. & Lee, R. J. Targeted delivery systems for oligonucleotide therapeutics. AAPS J. 11, 195–203 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Salvati, A. et al. Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface. Nature Nanotechnol. 8, 137–143 (2013).

    CAS  Google Scholar 

  109. Sahay, G. et al. Efficiency of siRNA delivery by lipid nanoparticles is limited by endocytic recycling. Nature Biotech. 31, 653–658 (2013). This study elucidates the mechanism of cellular uptake of LNPs and the importance of exocytosis in limiting delivery efficiency.

    CAS  Google Scholar 

  110. Schwarz, D. S. et al. Asymmetry in the assembly of the RNAi enzyme complex. Cell 115, 199–208 (2003).

    CAS  PubMed  Google Scholar 

  111. Geisbert, T. W. et al. Postexposure protection of non-human primates against a lethal Ebola virus challenge with RNA interference: a proof-of-concept study. Lancet 375, 1896–1905 (2010).

    CAS  PubMed  Google Scholar 

  112. Zimmermann, T. S. et al. RNAi-mediated gene silencing in non-human primates. Nature 441, 111–114 (2006).

    CAS  PubMed  Google Scholar 

  113. Wheeler, J. J. et al. Stabilized plasmid–lipid particles: construction and characterization. Gene Ther. 6, 271–281 (1999).

    CAS  PubMed  Google Scholar 

  114. Morrissey, D. V. et al. Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nature Biotech. 23, 1002–1007 (2005).

    CAS  Google Scholar 

  115. Jayaraman, M. et al. Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Angew. Chem. Int. Ed. Engl. 51, 8529–8533 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Akinc, A. et al. Development of lipidoid–siRNA formulations for systemic delivery to the liver. Mol. Ther. 17, 872–879 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Alabi, C. A. et al. Multiparametric approach for the evaluation of lipid nanoparticles for siRNA delivery. Proc. Natl Acad. Sci. USA 110, 12881–12886 (2013).

    CAS  PubMed  Google Scholar 

  118. Burnett, J. C., Rossi, J. J. & Tiemann, K. Current progress of siRNA/shRNA therapeutics in clinical trials. Biotechnol. J. 6, 1130–1146 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Fitzgerald, K. et al. Effect of an RNA interference drug on the synthesis of proprotein convertase subtilisin/kexin type 9 (PCSK9) and the concentration of serum LDL cholesterol in healthy volunteers: a randomised, single-blind, placebo-controlled, phase 1 trial. Lancet 383, 60–68 (2014).

    CAS  PubMed  Google Scholar 

  120. Santel, A. et al. A novel siRNA–lipoplex technology for RNA interference in the mouse vascular endothelium. Gene Ther. 13, 1222–1234 (2006).

    CAS  PubMed  Google Scholar 

  121. Santel, A. et al. RNA interference in the mouse vascular endothelium by systemic administration of siRNA–lipoplexes for cancer therapy. Gene Ther. 13, 1360–1370 (2006).

    CAS  PubMed  Google Scholar 

  122. Aleku, M. et al. Atu027, a liposomal small interfering RNA formulation targeting protein kinase N3, inhibits cancer progression. Cancer Res. 68, 9788–9798 (2008).

    CAS  PubMed  Google Scholar 

  123. Landen, C. N. Jr. et al. Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery. Cancer Res. 65, 6910–6918 (2005).

    CAS  PubMed  Google Scholar 

  124. Davis, M. E. et al. Self-assembling nucleic acid delivery vehicles via linear, water-soluble, cyclodextrin-containing polymers. Curr. Med. Chem. 11, 179–197 (2004).

    CAS  PubMed  Google Scholar 

  125. Hu-Lieskovan, S., Heidel, J. D., Bartlett, D. W., Davis, M. E. & Triche, T. J. Sequence-specific knockdown of EWS–FLI1 by targeted, nonviral delivery of small interfering RNA inhibits tumor growth in a murine model of metastatic Ewing's sarcoma. Cancer Res. 65, 8984–8992 (2005).

    CAS  PubMed  Google Scholar 

  126. Bartlett, D. W. & Davis, M. E. Impact of tumor-specific targeting and dosing schedule on tumor growth inhibition after intravenous administration of siRNA-containing nanoparticles. Biotechnol. Bioeng. 99, 975–985 (2008).

    CAS  PubMed  Google Scholar 

  127. Heidel, J. D. et al. Administration in non-human primates of escalating intravenous doses of targeted nanoparticles containing ribonucleotide reductase subunit M2 siRNA. Proc. Natl Acad. Sci. USA 104, 5715–5721 (2007).

    CAS  PubMed  Google Scholar 

  128. Davis, M. E. et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 464, 1067–1070 (2010). This paper provides the first evidence that systemically administered siRNA can induce RNAi in humans.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Wong, S. C. et al. Co-injection of a targeted, reversibly masked endosomolytic polymer dramatically improves the efficacy of cholesterol-conjugated small interfering RNAs in vivo. Nucleic Acid. Ther. 22, 380–390 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Rozema, D. B., Ekena, K., Lewis, D. L., Loomis, A. G. & Wolff, J. A. Endosomolysis by masking of a membrane-active agent (EMMA) for cytoplasmic release of macromolecules. Bioconjug.Chem. 14, 51–57 (2003).

    CAS  PubMed  Google Scholar 

  131. Wooddell, C. I. et al. Hepatocyte-targeted RNAi therapeutics for the treatment of chronic hepatitis B virus infection. Mol. Ther. 21, 973–985 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Yasuda, M. et al. RNAi-mediated silencing of hepatic Alas1 effectively prevents and treats the induced acute attacks in acute intermittent porphyria mice. Proc. Natl Acad. Sci. USA http://dx.doi.org/10.1073/pnas.1406228111 (2014).

  133. Chen, K. & Rajewsky, N. The evolution of gene regulation by transcription factors and microRNAs. Nature Rev. Genet. 8, 93–103 (2007).

    CAS  PubMed  Google Scholar 

  134. Pritchard, C. C., Cheng, H. H. & Tewari, M. MicroRNA profiling: approaches and considerations. Nature Rev. Genet. 13, 358–369 (2012).

    CAS  PubMed  Google Scholar 

  135. Hydbring, P., Badalian-Very, G. Clinical applications of microRNAs F1000Res. 2, 136 (2013).

    PubMed  PubMed Central  Google Scholar 

  136. Nana-Sinkam, S. P. & Croce, C. M. Clinical applications for microRNAs in cancer. Clin. Pharmacol. Ther. 93, 98–104 (2013).

    CAS  PubMed  Google Scholar 

  137. He, L. et al. A microRNA component of the p53 tumour suppressor network. Nature 447, 1130–1134 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Trang, P. et al. Systemic delivery of tumor suppressor microRNA mimics using a neutral lipid emulsion inhibits lung tumors in mice. Mol. Ther. 19, 1116–1122 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Bader, A. G. miR-34 — a microRNA replacement therapy is headed to the clinic. Front. Genet. 3, 120 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Gaj, T., Gersbach, C. A. & Barbas, C. F. 3rd. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 31, 397–405 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Urnov, F. D., Rebar, E. J., Holmes, M. C., Zhang, H. S. & Gregory, P. D. Genome editing with engineered zinc finger nucleases. Nature Rev. Genet. 11, 636–646 (2010).

    CAS  PubMed  Google Scholar 

  142. Boch, J. et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 326, 1509–1512 (2009).

    CAS  PubMed  Google Scholar 

  143. Moscou, M. J. & Bogdanove, A. J. A simple cipher governs DNA recognition by TAL effectors. Science 326, 1501 (2009).

    CAS  PubMed  Google Scholar 

  144. Miller, J. C. et al. A TALE nuclease architecture for efficient genome editing. Nature Biotech. 29, 143–148 (2011).

    CAS  Google Scholar 

  145. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Wang, H. et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153, 910–918 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Ran, F. A. et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell 154, 1380–1389 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Li, H. et al. In vivo genome editing restores haemostasis in a mouse model of haemophilia. Nature 475, 217–221 (2011). This study is the first to show that genome editing tools (ZFN) can modify gene in vivo and rescue disease phenotype.

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Yin, H. et al. Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nature Biotech. 32, 551–553 (2014). This paper is the first to show that the Cas9 system can correct a disease mutation and rescue disease phenotype in adult animals.

    CAS  Google Scholar 

  151. Hopkins, A. L. & Groom, C. R. The druggable genome. Nature Rev. Drug Discov. 1, 727–730 (2002).

    CAS  Google Scholar 

  152. Heyes, J., Palmer, L., Bremner, K. & MacLachlan, I. Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids. J. Control. Release 107, 276–287 (2005).

    CAS  PubMed  Google Scholar 

  153. Ambegia, E. et al. Stabilized plasmid-lipid particles containing PEG–diacylglycerols exhibit extended circulation lifetimes and tumor selective gene expression. Biochim. Biophys. Acta 1669, 155–163 (2005).

    CAS  PubMed  Google Scholar 

  154. Mishra, S., Heidel, J. D., Webster, P. & Davis, M. E. Imidazole groups on a linear, cyclodextrin-containing polycation produce enhanced gene delivery via multiple processes. J. Control. Release 116, 179–191 (2006).

    CAS  PubMed  Google Scholar 

  155. Bellocq, N. C., Pun, S. H., Jensen, G. S. & Davis, M. E. Transferrin-containing, cyclodextrin polymer-based particles for tumor-targeted gene delivery. Bioconjug. Chem. 14, 1122–1132 (2003).

    CAS  PubMed  Google Scholar 

  156. Mishra, S., Webster, P. & Davis, M. E. PEGylation significantly affects cellular uptake and intracellular trafficking of non-viral gene delivery particles. Eur. J. Cell Biol. 83, 97–111 (2004).

    CAS  PubMed  Google Scholar 

  157. Ramalingam, S., Annaluru, N. & Chandrasegaran, S. A CRISPR way to engineer the human genome. Genome Biol. 14, 107 (2013).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank R. L. Bogorad, E. A. Appel, O. F. Khan, O. Veiseh, Y. Dong and G. Sahay for discussion. H.Y. is supported by the US National Institutes of Health (NIH) Centers for Cancer Nanotechnology Excellence and the Harvard–MIT (Massachusetts Institute of Technology) Center of Cancer Nanotechnology Excellence (5-U54-CA151884-04). R.K. is supported by the US National Science Foundation Graduate Research Fellowship (grant 1122374). A.A.E. is supported by the US National Heart, Lung, and Blood Institute, NIH, as a Program of Excellence in Nanotechnology (PEN) Award (contract HHSN268201000045C). This work was supported partly by the Koch Institute Support (core) Grant P30-CA14051 from the US National Cancer Institute. The authors acknowledge the service to the MIT community of the late S. Collier.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniel G. Anderson.

Ethics declarations

Competing interests

D.G.A. is a cofounder of CRISPR Therapeutics. D.G.A. has a research grant with Alnylam Pharmaceuticals and a research grant with Shire. J.R.D. is a shareholder of Alnylam Pharmaceuticals.

Related links

PowerPoint slides

Glossary

Retroviruses

Single-stranded RNA viruses that use reverse transcriptase to transcribe their RNA into DNA, which can be integrated into the host genome. The viral DNA can be transcribed, translated and packed into new viruses. Replication-defective retroviruses are commonly used for gene therapy purposes. Most of these retroviruses are only active in dividing cells.

Lentiviruses

A subclass of retroviruses that are active in non-dividing cells. The replication-defective viruses are used both as a research tool to introduce a gene in vitro or in vivo and for gene therapy. They infect cells with high efficiency and introduce stable expression.

Adenoviruses

DNA viruses that do not integrate into the host genome and that usually do not replicate during cell division. Many of them trigger fast immune responses. For gene therapy purposes, they are usually applied in conditions in which temporary expression of proteins is required.

Adeno-associated viruses

(AAVs). DNA viruses that have very low but measurable genome integration rates and that are used as vectors for gene therapy. They are able to infect both dividing and non-dividing cells with high efficiency and long persistence. Although they have small packing capability, they are preferred for gene therapy owing to their low immunogenicity and low cytotoxicity.

Zinc-finger proteins

(ZFPs). DNA-binding proteins that consist of tandem arrays of zinc-fingers, which are protein structure motifs that contain one or more zinc ions. Engineered zinc-fingers have been shown to recognize three specific base pairs of DNA sequences and can be assembled in tandem to recognize specific nucleic acid sequences. The process of engineering is difficult and requires expertise.

Transcription activator-like effectors

(TALEs). Proteins that were first discovered in Xanthomonas spp. bacteria and that bind to promoter sequences in host plants to facilitate infections. They contain a repeat domain of 34 amino acids. Two critical amino acids in each repeat allow targeting of specific DNA bases. TALEs can be engineered in a time-consuming process by assembling repeat domains to recognize specific DNA sequences.

CRISPR–Cas

(Clustered regularly interspaced short palindromic repeat–CRISPR-associated). Defense systems against foreign DNA in bacteria and archaea, in which a short CRISPR RNA (crRNA) is used to guide the Cas nuclease to a specific target DNA sequence. These systems have been optimized to function in mammalian cells with high efficiency. The engineering process to target various DNA sequences is straightforward, and the cost is low.

Liposomes

Vesicles of various sizes with a lipid bilayer that can encapsulate small molecules or large molecules such as small interfering RNA or DNA. By fusing to the cell membrane, they facilitate delivery of the liposome contents in vitro and in vivo.

Polymersomes

Vesicles produced using co-polymers, which are made of two or more monomers, to allow both hydrophilic and lipophilic ability. They can encapsulate small molecules, proteins and DNA to form particles of various sizes, and are used for drug delivery systems in vitro and in vivo.

Cell-penetrating peptides

Small peptides that can translocate across plasma membranes. By covalent or non-covalent binding of these peptides to small molecules, proteins, DNA, small interfering RNA or even nanoparticles, they could facilitate intracellular delivery of various types of molecules.

Scaffold/matrix attachment regions

(S/MARs). Anchor points of the genomic DNA for the chromatin scaffold. They are found at introns or borders of transcription units, where they have an important role in separating these units and regulating gene expression.

PhiC31

A sequence-specific recombinase that mediates recombination between 2 specific 34-bp sequences, which allows insertion of DNA into another DNA molecule or into the genome at specific sites.

PiggyBac

A transposon system composed of a transposon and a transposase, which recognizes transposon-specific sequences on both ends of a vector and integrates the content into different DNA molecules or genome.

Sleeping Beauty

A transposon system reconstructed from DNA copies of salmon. The transposase was engineered to facilitate robust and stable gene transfer.

RNAiMAX

A commercial reagent that delivers small interfering RNAs into various types of cells in vitro with high efficiency. As a cationic lipid formulation, it can also be used to deliver microRNA antagonists and mimics, as well as mRNAs.

Glomerular filtration barrier

A blood filtration interface in the kidney that allows free passage of small ions such as sodium and potassium but that retains large proteins. The cutoff to pass this barrier is ~70 kDa, and naked small interfering RNAs (~13 kDa) can thus be filtered through the kidney.

FokI

A restriction endonuclease with a DNA-binding domain at the amino terminus and a nonspecific DNA cleavage domain at the carboxyl terminus. Dimerization of two FokI is required to cleave DNA. When it is fused to zinc-finger proteins (ZFPs) or to transcription activator-like effectors (TALEs), FokI will function when a pair of ZFPs or TALEs binds to DNA within short distance. This strategy allows increased specificity of sequence recognition.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yin, H., Kanasty, R., Eltoukhy, A. et al. Non-viral vectors for gene-based therapy. Nat Rev Genet 15, 541–555 (2014). https://doi.org/10.1038/nrg3763

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3763

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research