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.

  • Review Article
  • Published:

Antisense oligonucleotides: the next frontier for treatment of neurological disorders

Key Points

  • Antisense oligonucleotides (ASOs) are short, synthetic, single-stranded oligodeoxynucleotides that can alter RNA and reduce, restore, or modify protein expression through several distinct mechanisms

  • By targeting the source of the pathogenesis, ASO-mediated therapies have an higher chance of success than therapies targeting downstream pathways

  • Advances in the understanding of ASO pharmacology have provided momentum for translating these therapeutics into the clinic

  • Two ASO-mediated therapies have received approval from the US Food and Drug Administration for the treatment of Duchenne muscular dystrophy and spinal muscular atrophy

  • Further advancement of ASOs in the clinic urgently requires optimization of ASO delivery, target engagement, and safety profile

  • This technology holds the potential to change the therapeutic landscape for many neurological and non-neurological conditions in the near future

Abstract

Antisense oligonucleotides (ASOs) were first discovered to influence RNA processing and modulate protein expression over two decades ago; however, progress translating these agents into the clinic has been hampered by inadequate target engagement, insufficient biological activity, and off-target toxic effects. Over the years, novel chemical modifications of ASOs have been employed to address these issues. These modifications, in combination with elucidation of the mechanism of action of ASOs and improved clinical trial design, have provided momentum for the translation of ASO-based strategies into therapies. Many neurological conditions lack an effective treatment; however, as research progressively disentangles the pathogenic mechanisms of these diseases, they provide an ideal platform to test ASO-based strategies. This steady progress reached a pinnacle in the past few years with approvals of ASOs for the treatment of spinal muscular atrophy and Duchenne muscular dystrophy, which represent landmarks in a field in which disease-modifying therapies were virtually non-existent. With the rapid development of improved next-generation ASOs toward clinical application, this technology now holds the potential to have a dramatic effect on the treatment of many neurological conditions in the near future.

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: Chemical modifications of the ASO backbone.
Figure 2: Functional mechanisms of ASOs.

Similar content being viewed by others

References

  1. Stephenson, M. L. & Zamecnik, P. C. Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proc. Natl Acad. Sci. USA 75, 285–288 (1978).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Muntoni, F. & Wood, M. J. A. Targeting RNA to treat neuromuscular disease. Nat. Rev. Drug Discov. 10, 621–637 (2011).

    Article  CAS  PubMed  Google Scholar 

  3. Opalinska, J. B. & Gewirtz, A. M. Nucleic-acid therapeutics: basic principles and recent applications. Nat. Rev. Drug Discov. 1, 503–514 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Juliano, R. L. The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 44, 6518–6548 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Dias, N. & Stein, C. A. Antisense oligonucleotides: basic concepts and mechanisms. Mol. Cancer Ther. 1, 347–355 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Eder, P. S., DeVine, R. J., Dagle, J. M. & Walder, J. A. Substrate specificity and kinetics of degradation of antisense oligonucleotides by a 3′ exonuclease in plasma. Antisense Res. Dev. 1, 141–151 (1991).

    Article  CAS  PubMed  Google Scholar 

  7. De Clercq, E., Eckstein, E. & Merigan, T. C. Interferon induction increased through chemical modification of a synthetic polyribonucleotide. Science 165, 1137–1139 (1969).

    Article  CAS  PubMed  Google Scholar 

  8. Rifai, A., Brysch, W., Fadden, K., Clark, J. & Schlingensiepen, K. H. Clearance kinetics, biodistribution, and organ saturability of phosphorothioate oligodeoxynucleotides in mice. Am. J. Pathol. 149, 717–725 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Watanabe, T. A., Geary, R. S. & Levin, A. A. Plasma protein binding of an antisense oligonucleotide targeting human ICAM-1 (ISIS 2302). Oligonucleotides 16, 169–180 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Wu, H. et al. Determination of the role of the human RNase H1 in the pharmacology of DNA-like antisense drugs. J. Biol. Chem. 279, 17181–17189 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Freier, S. M. & Altmann, K. H. The ups and downs of nucleic acid duplex stability: structure-stability studies on chemically-modified DNA:RNA duplexes. Nucleic Acids Res. 25, 4429–4443 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lubini, P., Zürcher, W. & Egli, M. Stabilizing effects of the RNA 2′-substituent: crystal structure of an oligodeoxynucleotide duplex containing 2′-O-methylated adenosines. Chem. Biol. 1, 39–45 (1994).

    Article  CAS  PubMed  Google Scholar 

  13. McKay, R. A. et al. Characterization of a potent and specific class of antisense oligonucleotide inhibitor of human protein kinase C-alpha expression. J. Biol. Chem. 274, 1715–1722 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Geary, R. S., Yu, R. Z. & Levin, A. A. Pharmacokinetics of phosphorothioate antisense oligodeoxynucleotides. Curr. Opin. Investig. Drugs 2, 562–573 (2001).

    CAS  PubMed  Google Scholar 

  15. Hamm, S. et al. Alternating 2′-O-ribose methylation is a universal approach for generating non-stimulatory siRNA by acting as TLR7 antagonist. Immunobiology 215, 559–569 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Summerton, J. et al. Morpholino and phosphorothioate antisense oligomers compared in cell-free and in-cell systems. Antisense Nucleic Acid Drug Dev. 7, 63–70 (1997).

    Article  CAS  PubMed  Google Scholar 

  17. Hudziak, R. M. et al. Resistance of morpholino phosphorodiamidate oligomers to enzymatic degradation. Antisense Nucleic Acid Drug Dev. 6, 267–272 (1996).

    Article  CAS  PubMed  Google Scholar 

  18. Geary, R. S. Antisense oligonucleotide pharmacokinetics and metabolism. Expert Opin. Drug Metab. Toxicol. 5, 381–391 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. Yu, R. Z., Grundy, J. S. & Geary, R. S. Clinical pharmacokinetics of second generation antisense oligonucleotides. Expert Opin. Drug Metab. Toxicol. 9, 169–182 (2013).

    Article  CAS  PubMed  Google Scholar 

  20. Crooke, S. T. & Geary, R. S. Clinical pharmacological properties of mipomersen (Kynamro), a second generation antisense inhibitor of apolipoprotein B. Br. J. Clin. Pharmacol. 76, 269–276 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Levin, A. A., Levine, M. S., Rubesin, S. E. & Laufer, I. An 8-year review of barium studies in the diagnosis of gastroparesis. Clin. Radiol. 63, 407–414 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Amantana, A. & Iversen, P. L. Pharmacokinetics and biodistribution of phosphorodiamidate morpholino antisense oligomers. Curr. Opin. Pharmacol. 5, 550–555 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Thompson, J. D. et al. Toxicological and pharmacokinetic properties of chemically modified siRNAs targeting p53 RNA following intravenous administration. Nucleic Acid Ther. 22, 255–264 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yu, R. Z. et al. Cross-species comparison of in vivo PK/PD relationships for second-generation antisense oligonucleotides targeting apolipoprotein B-100. Biochem. Pharmacol. 77, 910–919 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Zhang, H. et al. Reduction of liver Fas expression by an antisense oligonucleotide protects mice from fulminant hepatitis. Nat. Biotechnol. 18, 862–867 (2000).

    Article  CAS  PubMed  Google Scholar 

  26. Altmann, K. H. et al. Second-generation antisense oligonucleotides: structure-activity relationships and the design of improved signal-transduction inhibitors. Biochem. Soc. Trans. 24, 630–637 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. Hung, G. et al. Characterization of target mRNA reduction through in situ RNA hybridization in multiple organ systems following systemic antisense treatment in animals. Nucleic Acid Ther. 23, 369–378 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Phillips, J. A. et al. Pharmacokinetics, metabolism, and elimination of a 20-mer phosphorothioate oligodeoxynucleotide (CGP 69846A) after intravenous and subcutaneous administration. Biochem. Pharmacol. 54, 657–668 (1997).

    Article  CAS  PubMed  Google Scholar 

  29. Smith, R. A. et al. Antisense oligonucleotide therapy for neurodegenerative disease. J. Clin. Invest. 116, 2290–2296 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kordasiewicz, H. B. et al. Sustained therapeutic reversal of Huntington's Disease by transient repression of Huntingtin synthesis. Neuron 74, 1031–1044 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Passini, M. A. et al. Antisense oligonucleotides delivered to the mouse CNS ameliorate symptoms of severe spinal muscular atrophy. Sci. Transl Med. 3, 72ra18 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Rigo, F. et al. Pharmacology of a central nervous system delivered 2′-O-methoxyethyl-modified survival of motor neuron splicing oligonucleotide in mice and nonhuman primates. J. Pharmacol. Exp. Ther. 350, 46–55 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Southwell, A. L., Skotte, N. H., Bennett, C. F. & Hayden, M. R. Antisense oligonucleotide therapeutics for inherited neurodegenerative diseases. Trends Mol. Med. 18, 634–643 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Finkel, R. S. et al. Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study. Lancet 388, 3017–3026 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Miller, T. M. et al. An antisense oligonucleotide against SOD1 delivered intrathecally for patients with SOD1 familial amyotrophic lateral sclerosis: a phase 1, randomised, first-in-man study. Lancet Neurol. 12, 435–442 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ostergaard, M. E. et al. Rational design of antisense oligonucleotides targeting single nucleotide polymorphisms for potent and allele selective suppression of mutant Huntingtin in the CNS. Nucleic Acids Res. 41, 9634–9650 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chiriboga, C. A. et al. Results from a phase 1 study of nusinersen (ISIS-SMNRx) in children with spinal muscular atrophy. Neurology 86, 890–897 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Alter, J. et al. Systemic delivery of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic pathology. Nat. Med. 12, 175–177 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Benimetskaya, L. et al. Mac-1 (CD11b/CD18) is an oligodeoxynucleotide-binding protein. Nat. Med. 3, 414–420 (1997).

    Article  CAS  PubMed  Google Scholar 

  40. Butler, M., Stecker, K. & Bennett, C. F. Cellular distribution of phosphorothioate oligodeoxynucleotides in normal rodent tissues. Lab. Invest. 77, 379–388 (1997).

    CAS  PubMed  Google Scholar 

  41. Robbins, M., Judge, A. & MacLachlan, I. siRNA and innate immunity. Oligonucleotides 19, 89–102 (2009).

    Article  CAS  PubMed  Google Scholar 

  42. Kawai, T. & Akira, S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 34, 637–650 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Kortylewski, M. et al. In vivo delivery of siRNA to immune cells by conjugation to a TLR9 agonist enhances antitumor immune responses. Nat. Biotechnol. 27, 925–932 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lorenz, P., Misteli, T., Baker, B. F., Bennett, C. F. & Spector, D. L. Nucleocytoplasmic shuttling: a novel in vivo property of antisense phosphorothioate oligodeoxynucleotides. Nucleic Acids Res. 28, 582–592 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hartig, R., Shoeman, R. L., Janetzko, A., Grüb, S. & Traub, P. Active nuclear import of single-stranded oligonucleotides and their complexes with non-karyophilic macromolecules. Biol. Cell 90, 407–426 (1998).

    Article  CAS  PubMed  Google Scholar 

  46. Varkouhi, A. K., Scholte, M., Storm, G. & Haisma, H. J. Endosomal escape pathways for delivery of biologicals. J. Control. Release 151, 220–228 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Juliano, R. L., Ming, X. & Nakagawa, O. Cellular uptake and intracellular trafficking of antisense and siRNA oligonucleotides. Bioconjug. Chem. 23, 147–157 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Juliano, R. L. & Carver, K. Cellular uptake and intracellular trafficking of oligonucleotides. Adv. Drug Deliv. Rev. 87, 35–45 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wagenaar, T. R. et al. Identification of the endosomal sorting complex required for transport-I (ESCRT-I) as an important modulator of anti-miR uptake by cancer cells. Nucleic Acids Res. 43, 1204–1215 (2015).

    Article  CAS  PubMed  Google Scholar 

  50. Tangsangasaksri, M. et al. siRNA-loaded polyion complex micelle decorated with charge-conversional polymer tuned to undergo stepwise response to intra-tumoral and intra-endosomal pHs for exerting enhanced RNAi efficacy. Biomacromolecules 17, 246–255 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Yang, B. et al. High-throughput screening identifies small molecules that enhance the pharmacological effects of oligonucleotides. Nucleic Acids Res. 43, 1987–1996 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Majorek, K. A. et al. The RNase H-like superfamily: new members, comparative structural analysis and evolutionary classification. Nucleic Acids Res. 42, 4160–4179 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lima, W. F., De Hoyos, C. L., Liang, X. & Crooke, S. T. RNA cleavage products generated by antisense oligonucleotides and siRNAs are processed by the RNA surveillance machinery. Nucleic Acids Res. 44, 3351–3363 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Deleavey, G. F. & Damha, M. J. Designing chemically modified oligonucleotides for targeted gene silencing. Chem. Biol. 19, 937–954 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Monia, B. P. et al. Evaluation of 2′-modified oligonucleotides containing 2′-deoxy gaps as antisense inhibitors of gene expression. J. Biol. Chem. 268, 14514–14522 (1993).

    CAS  PubMed  Google Scholar 

  56. Wu, H., Lima, W. F. & Crooke, S. T. Properties of cloned and expressed human RNase H1. J. Biol. Chem. 274, 28270–28278 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. Breaker, R. R. & Joyce, G. F. A. DNA enzyme that cleaves RNA. Chem. Biol. 1, 223–229 (1994).

    Article  CAS  PubMed  Google Scholar 

  58. Cech, T. R. & Uhlenbeck, O. C. Hammerhead nailed down. Nature 372, 39–40 (1994).

    Article  CAS  PubMed  Google Scholar 

  59. Baker, B. F. et al. 2′-O-(2-Methoxy)ethyl-modified anti-intercellular adhesion molecule 1 (ICAM-1) oligonucleotides selectively increase the ICAM-1 mRNA level and inhibit formation of the ICAM-1 translation initiation complex in human umbilical vein endothelial cells. J. Biol. Chem. 272, 11994–12000 (1997).

    Article  CAS  PubMed  Google Scholar 

  60. Evers, M. M., Toonen, L. J. A. & van Roon-Mom, W. M. C. Antisense oligonucleotides in therapy for neurodegenerative disorders. Adv. Drug Deliv. Rev. 87, 90–103 (2015).

    Article  CAS  PubMed  Google Scholar 

  61. Havens, M. A. & Hastings, M. L. SURVEY AND SUMMARY Splice-switching antisense oligonucleotides as therapeutic drugs. Nucleic Acids Res. 44, 6549–6563 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  62. US Food and Drug Administration. FDA approves first drug for spinal muscular atrophy. US Food and Drug Administration https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm534611.htm (2016).

  63. Vickers, T. A., Wyatt, J. R., Burckin, T., Bennett, C. F. & Freier, S. M. Fully modified 2' MOE oligonucleotides redirect polyadenylation. Nucleic Acids Res. 29, 1293–1299 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Dominski, Z. & Kole, R. Restoration of correct splicing in thalassemic pre-mRNA by antisense oligonucleotides. Proc. Natl Acad. Sci. USA 90, 8673–8677 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Mah, J. K. et al. A systematic review and meta-analysis on the epidemiology of Duchenne and Becker muscular dystrophy. Neuromuscul. Disord. 24, 482–491 (2014).

    Article  PubMed  Google Scholar 

  66. Judge, L. M., Haraguchiln, M. & Chamberlain, J. S. Dissecting the signaling and mechanical functions of the dystrophin-glycoprotein complex. J. Cell Sci. 119, 1537–1546 (2006).

    Article  CAS  PubMed  Google Scholar 

  67. Parker, A. E. et al. Analysis of an adult Duchenne muscular dystrophy population. QJM 98, 729–736 (2005).

    Article  CAS  PubMed  Google Scholar 

  68. Guiraud, S. et al. The pathogenesis and therapy of muscular dystrophies. Annu. Rev. Genomics Hum. Genet. 16, 281–308 (2015).

    Article  CAS  PubMed  Google Scholar 

  69. Davies, K. E. & Nowak, K. J. Molecular mechanisms of muscular dystrophies: old and new players. Nat. Rev. Mol. Cell Biol. 7, 762–773 (2006).

    Article  CAS  PubMed  Google Scholar 

  70. Hoffman, E. P., Brown, R. H. & Kunkel, L. M. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 51, 919–928 (1987).

    Article  CAS  PubMed  Google Scholar 

  71. Bladen, C. L. et al. The TREAT-NMD DMD global database: analysis of more than 7,000 Duchenne muscular dystrophy mutations. Hum. Mutat. 36, 395–402 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Dunckley, M. G., Manoharan, M., Villiet, P., Eperon, I. C. & Dickson, G. Modification of splicing in the dystrophin gene in cultured Mdx muscle cells by antisense oligoribonucleotides. Hum. Mol. Genet. 7, 1083–1090 (1998).

    Article  CAS  PubMed  Google Scholar 

  73. Pramono, Z. A. et al. Induction of exon skipping of the dystrophin transcript in lymphoblastoid cells by transfecting an antisense oligodeoxynucleotide complementary to an exon recognition sequence. Biochem. Biophys. Res. Commun. 226, 445–449 (1996).

    Article  CAS  PubMed  Google Scholar 

  74. Takeshima, Y., Nishio, H., Sakamoto, H., Nakamura, H. & Matsuo, M. Modulation of in vitro splicing of the upstream intron by modifying an intra-exon sequence which is deleted from the dystrophin gene in dystrophin Kobe. J. Clin. Invest. 95, 515–520 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Aartsma-Rus, A. et al. Targeted exon skipping as a potential gene correction therapy for Duchenne muscular dystrophy. Neuromuscul. Disord. 12 (Suppl. 1), S71–S77 (2002).

    Article  PubMed  Google Scholar 

  76. Aartsma-Rus, A. et al. Comparative analysis of antisense oligonucleotide analogs for targeted DMD exon 46 skipping in muscle cells. Gene Ther. 11, 1391–1398 (2004).

    Article  CAS  PubMed  Google Scholar 

  77. Popplewell, L. J. et al. Comparative analysis of antisense oligonucleotide sequences targeting exon 53 of the human DMD gene: Implications for future clinical trials. Neuromuscul. Disord. 20, 102–110 (2010).

    Article  PubMed  Google Scholar 

  78. Popplewell, L. J., Trollet, C., Dickson, G. & Graham, I. R. Design of phosphorodiamidate morpholino oligomers (PMOs) for the induction of exon skipping of the human DMD gene. Mol. Ther. 17, 554–561 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. van Deutekom, J. C. et al. Antisense-induced exon skipping restores dystrophin expression in DMD patient derived muscle cells. Hum. Mol. Genet. 10, 1547–1554 (2001).

    Article  CAS  PubMed  Google Scholar 

  80. van Deutekom, J. C. T. & van Ommen, G.-J. B. Advances in Duchenne muscular dystrophy gene therapy. Nat. Rev. Genet. 4, 774–783 (2003).

    Article  CAS  PubMed  Google Scholar 

  81. Yokota, T., Duddy, W. & Partridge, T. Optimizing exon skipping therapies for DMD. Acta Myol. 26, 179–184 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Lu, Q. L. et al. Systemic delivery of antisense oligoribonucleotide restores dystrophin expression in body-wide skeletal muscles. Proc. Natl Acad. Sci. USA 102, 198–203 (2005).

    Article  CAS  PubMed  Google Scholar 

  83. Heemskerk, H. et al. Preclinical PK and PD studies on 2′-O-methyl-phosphorothioate RNA antisense oligonucleotides in the mdx mouse model. Mol. Ther. 18, 1210–1217 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Heemskerk, H. A. et al. In vivo comparison of 2′-O-methyl phosphorothioate and morpholino antisense oligonucleotides for Duchenne muscular dystrophy exon skipping. J. Gene Med. 11, 257–266 (2009).

    Article  CAS  PubMed  Google Scholar 

  85. Yokota, T., Hoffman, E. & Takeda, S. Antisense oligo-mediated multiple exon skipping in a dog model of duchenne muscular dystrophy. Methods Mol. Biol. 709, 299–312 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Malerba, A., Boldrin, L. & Dickson, G. Long-term systemic administration of unconjugated morpholino oligomers for therapeutic expression of dystrophin by exon skipping in skeletal muscle: implications for cardiac muscle integrity. Nucleic Acid. Ther. 21, 293–298 (2011).

    Article  CAS  PubMed  Google Scholar 

  87. Townsend, D., Yasuda, S., Li, S., Chamberlain, J. S. & Metzger, J. M. Emergent dilated cardiomyopathy caused by targeted repair of dystrophic skeletal muscle. Mol. Ther. 16, 832–835 (2008).

    Article  CAS  PubMed  Google Scholar 

  88. Cirak, S. et al. Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. Lancet 378, 595–605 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Mendell, J. R. et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann. Neurol. 74, 637–647 (2013).

    Article  CAS  PubMed  Google Scholar 

  90. Sarepta Therapeutics, Inc. PCNSD Advisory Committee Meeting Briefing Document. U.S. Food and Drug Administration https://www.fda.gov/downloads/advisorycommittees/committeesmeetingmaterials/drugs/peripheralandcentralnervoussystemdrugsadvisorycommittee/ucm497064.pdf (2017).

  91. US Food and Drug Administration. FDA grants accelerated approval to first drug for Duchenne muscular dystrophy. US Food and Drug Administration https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm521263.htm (2016).

  92. Kesselheim, A. S. & Avorn, J. Approving a problematic muscular dystrophy drug. JAMA 316, 2357 (2016).

    Article  PubMed  Google Scholar 

  93. Pearson, S., Jia, H. & Kandachi, K. China approves first gene therapy. Nat. Biotechnol. 22, 3–4 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Hoyng, S. A. et al. Gene therapy and peripheral nerve repair: a perspective. Front. Mol. Neurosci. 8, 32 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Leader, B., Baca, Q. J. & Golan, D. E. Protein therapeutics: a summary and pharmacological classification. Nat. Rev. Drug Discov. 7, 21–39 (2008).

    Article  CAS  PubMed  Google Scholar 

  96. Liang, X. et al. Translation efficiency of mRNAs is increased by antisense oligonucleotides targeting upstream open reading frames. Nat. Biotechnol. 34, 875–880 (2016).

    Article  CAS  PubMed  Google Scholar 

  97. Calvo, S. E., Pagliarini, D. J. & Mootha, V. K. Upstream open reading frames cause widespread reduction of protein expression and are polymorphic among humans. Proc. Natl Acad. Sci. USA 106, 7507–7512 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  98. Lee, S. et al. Global mapping of translation initiation sites in mammalian cells at single-nucleotide resolution. Proc. Natl Acad. Sci. USA 109, E2424–E2432 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Orr, H. T. & Zoghbi, H. Y. Trinucleotide repeat disorders. Annu. Rev. Neurosci. 30, 575–621 (2007).

    Article  CAS  PubMed  Google Scholar 

  100. Davies, S. W. et al. Formation of neuronal intranuclear inclusions underlies the neurological dysfunction in mice transgenic for the HD mutation. Cell 90, 537–548 (1997).

    Article  CAS  PubMed  Google Scholar 

  101. Seidel, K. et al. Brain pathology of spinocerebellar ataxias. Acta Neuropathol. 124, 1–21 (2012).

    Article  CAS  PubMed  Google Scholar 

  102. Hu, J. et al. Allele-specific silencing of mutant huntingtin and ataxin-3 genes by targeting expanded CAG repeats in mRNAs. Nat. Biotechnol. 27, 478–484 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Hu, J., Liu, J. & Corey, D. R. Allele-selective inhibition of Huntingtin expression by switching to an miRNA-like RNAi mechanism. Chem. Biol. 17, 1183–1188 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Gagnon, K. T. et al. Allele-selective inhibition of mutant Huntingtin expression with antisense oligonucleotides targeting the expanded CAG repeat. Biochemistry 49, 10166–10178 (2010).

    Article  CAS  PubMed  Google Scholar 

  105. Sun, X. et al. Phosphorodiamidate morpholino oligomers suppress mutant huntingtin expression and attenuate neurotoxicity. Hum. Mol. Genet. 23, 6302–6317 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. van Bilsen, P. H. J. et al. Identification and allele-specific silencing of the mutant huntingtin allele in Huntington's disease patient-derived fibroblasts. Hum. Gene Ther. 19, 710–718 (2008).

    Article  CAS  PubMed  Google Scholar 

  107. Lombardi, M. S. et al. A majority of Huntington's disease patients may be treatable by individualized allele-specific RNA interference. Exp. Neurol. 217, 312–319 (2009).

    Article  CAS  PubMed  Google Scholar 

  108. Carroll, J. B. et al. Potent and selective antisense oligonucleotides targeting single-nucleotide polymorphisms in the Huntington disease gene/ allele-specific silencing of mutant huntingtin. Mol. Ther. 19, 2178–2185 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Warby, S. C. et al. CAG expansion in the Huntington disease gene is associated with a specific and targetable predisposing haplogroup. Am. J. Hum. Genet. 84, 351–366 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Pfister, E. L. et al. Five siRNAs targeting three SNPs may provide therapy for three-quarters of Huntington's disease patients. Curr. Biol. 19, 774–778 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Rosen, D. R. et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 362, 59–62 (1993).

    Article  CAS  PubMed  Google Scholar 

  112. Winer, L. et al. SOD1 in cerebral spinal fluid as a pharmacodynamic marker for antisense oligonucleotide therapy. JAMA Neurol. 70, 201–207 (2013).

    Article  PubMed  Google Scholar 

  113. Crisp, M. J. et al. In vivo kinetic approach reveals slow SOD1 turnover in the CNS. J. Clin. Invest. 125, 2772–2780 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Andreadis, A. Misregulation of tau alternative splicing in neurodegeneration and dementia. Prog. Mol. Subcell. Biol. 44, 89–107 (2006).

    Article  CAS  PubMed  Google Scholar 

  115. Lee, G., Cowan, N. & Kirschner, M. The primary structure and heterogeneity of tau protein from mouse brain. Science 239, 285–288 (1988).

    Article  CAS  PubMed  Google Scholar 

  116. Lindwall, G. & Cole, R. D. Phosphorylation affects the ability of tau protein to promote microtubule assembly. J. Biol. Chem. 259, 5301–5305 (1984).

    CAS  PubMed  Google Scholar 

  117. Alonso, A. C., Zaidi, T., Grundke-Iqbal, I. & Iqbal, K. Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease. Proc. Natl Acad. Sci. USA 91, 5562–5566 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Grundke-Iqbal, I. et al. Abnormal phosphorylation of the microtubule-associated protein τ (tau) in Alzheimer cytoskeletal pathology. Proc. Natl Acad. Sci. USA 83, 4913–4917 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Iqbal, K. et al. Defective brain microtubule assembly in Alzheimer's disease. Lancet 2, 421–426 (1986).

    Article  CAS  PubMed  Google Scholar 

  120. Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259 (1991).

    Article  CAS  PubMed  Google Scholar 

  121. Tomlinson, B. E., Blessed, G. & Roth, M. Observations on the brains of demented old people. J. Neurol. Sci. 11, 205–242 (1970).

    Article  CAS  PubMed  Google Scholar 

  122. Alafuzoff, I., Iqbal, K., Friden, H., Adolfsson, R. & Winblad, B. Histopathological criteria for progressive dementia disorders: clinical-pathological correlation and classification by multivariate data analysis. Acta Neuropathol. 74, 209–225 (1987).

    Article  CAS  PubMed  Google Scholar 

  123. Arriagada, P. V., Growdon, J. H., Hedley-Whyte, E. T. & Hyman, B. T. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer's disease. Neurology 42, 631–639 (1992).

    Article  CAS  PubMed  Google Scholar 

  124. Hyman, B. T., Van Hoesen, G. W., Damasio, A. R. & Barnes, C. L. Alzheimer's disease: cell-specific pathology isolates the hippocampal formation. Science 225, 1168–1170 (1984).

    Article  CAS  PubMed  Google Scholar 

  125. Lei, P. et al. Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export. Nat. Med. 18, 291–295 (2012).

    CAS  PubMed  Google Scholar 

  126. Morris, M. et al. Age-appropriate cognition and subtle dopamine-independent motor deficits in aged Tau knockout mice. Neurobiol. Aging 34, 1523–1529 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Li, Z., Hall, A. M., Kelinske, M. & Roberson, E. D. Seizure resistance without parkinsonism in aged mice after tau reduction. Neurobiol. Aging 35, 2617–2624 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  128. Roberson, E. D. et al. Reducing endogenous tau ameliorates amyloid β-induced deficits in an Alzheimer's disease mouse model. Science 316, 750–754 (2007).

    Article  CAS  PubMed  Google Scholar 

  129. van Hummel, A. et al. No overt deficits in aged tau-deficient C57Bl/6.Mapttm1(EGFP)Kit GFP knockin mice. PLoS ONE 11, e0163236 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. DeVos, S. L. & Miller, T. M. Antisense oligonucleotides: treating neurodegeneration at the level of RNA. Neurotherapeutics 10, 486–497 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Sud, R., Geller, E. T. & Schellenberg, G. D. Antisense-mediated exon skipping decreases tau protein expression: a potential therapy for tauopathies. Mol. Ther. Nucleic Acids 3, e180 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Yoshiyama, Y. et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 53, 337–351 (2007).

    Article  CAS  PubMed  Google Scholar 

  133. DeVos, S. L. et al. Tau reduction prevents neuronal loss and reverses pathological tau deposition and seeding in mice with tauopathy. Sci. Transl Med. 9, eaag0481 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Schoch, K. M. et al. Increased 4R-tau induces pathological changes in a human-tau mouse model. Neuron 90, 941–947 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Jiang, H., Mankodi, A., Swanson, M. S., Moxley, R. T. & Thornton, C. A. Myotonic dystrophy type 1 is associated with nuclear foci of mutant RNA, sequestration of muscleblind proteins and deregulated alternative splicing in neurons. Hum. Mol. Genet. 13, 3079–3088 (2004).

    Article  CAS  PubMed  Google Scholar 

  136. Mahadevan, M. S. et al. Reversible model of RNA toxicity and cardiac conduction defects in myotonic dystrophy. Nat. Genet. 38, 1066–1070 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Ranum, L. P. W. & Cooper, T. A. RNA-mediated neuromuscular disorders. Annu. Rev. Neurosci. 29, 259–277 (2006).

    Article  CAS  PubMed  Google Scholar 

  138. Orengo, J. P. et al. Expanded CTG repeats within the DMPK 3′ UTR causes severe skeletal muscle wasting in an inducible mouse model for myotonic dystrophy. Proc. Natl Acad. Sci. USA 105, 2646–2651 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  139. Yadava, R. S. et al. RNA toxicity in myotonic muscular dystrophy induces NKX2-5 expression. Nat. Genet. 40, 61–68 (2008).

    Article  CAS  PubMed  Google Scholar 

  140. Day, J. W. et al. Myotonic dystrophy type 2: molecular, diagnostic and clinical spectrum. Neurology 60, 657–664 (2003).

    Article  CAS  PubMed  Google Scholar 

  141. Liquori, C. L. et al. Myotonic dystrophy Type 2: human founder haplotype and evolutionary conservation of the repeat tract. Am. J. Hum. Genet. 73, 849–862 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Margolis, J. M., Schoser, B. G., Moseley, M. L., Day, J. W. & Ranum, L. P. W. DM2 intronic expansions: evidence for CCUG accumulation without flanking sequence or effects on ZNF9 mRNA processing or protein expression. Hum. Mol. Genet. 15, 1808–1815 (2006).

    Article  CAS  PubMed  Google Scholar 

  143. Lee, J. E. & Cooper, T. A. Pathogenic mechanisms of myotonic dystrophy. Biochem. Soc. Trans. 37, 1281–1286 (2009).

    Article  CAS  PubMed  Google Scholar 

  144. Napierała, M. & Krzyzosiak, W. J. CUG repeats present in myotonin kinase RNA form metastable “slippery” hairpins. J. Biol. Chem. 272, 31079–31085 (1997).

    Article  PubMed  Google Scholar 

  145. Kuyumcu-Martinez, N. M. & Cooper, T. A. Misregulation of alternative splicing causes pathogenesis in myotonic dystrophy. Prog. Mol. Subcell. Biol. 44, 133–159 (2006).

    Article  CAS  PubMed  Google Scholar 

  146. Cardani, R., Mancinelli, E., Rotondo, G., Sansone, V. & Meola, G. Muscleblind-like protein 1 nuclear sequestration is a molecular pathology marker of DM1 and DM2. Eur. J. Histochem. 50, 177–182 (2006).

    CAS  PubMed  Google Scholar 

  147. Fardaei, M., Larkin, K., Brook, J. D. & Hamshere, M. G. In vivo co-localisation of MBNL protein with DMPK expanded-repeat transcripts. Nucleic Acids Res. 29, 2766–2771 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Fardaei, M. et al. Three proteins, MBNL, MBLL and MBXL, co-localize in vivo with nuclear foci of expanded-repeat transcripts in DM1 and DM2 cells. Hum. Mol. Genet. 11, 805–814 (2002).

    CAS  Google Scholar 

  149. Ward, A. J., Rimer, M., Killian, J. M., Dowling, J. J. & Cooper, T. A. CUGBP1 overexpression in mouse skeletal muscle reproduces features of myotonic dystrophy type 1. Hum. Mol. Genet. 19, 3614–3622 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Yuan, Y. et al. Muscleblind-like 1 interacts with RNA hairpins in splicing target and pathogenic RNAs. Nucleic Acids Res. 35, 5474–5486 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Koshelev, M., Sarma, S., Price, R. E., Wehrens, X. H. T. & Cooper, T. A. Heart-specific overexpression of CUGBP1 reproduces functional and molecular abnormalities of myotonic dystrophy type 1. Hum. Mol. Genet. 19, 1066–1075 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Du, H. et al. Aberrant alternative splicing and extracellular matrix gene expression in mouse models of myotonic dystrophy. Nat. Struct. Mol. Biol. 17, 187–193 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Wheeler, T. M. et al. Reversal of RNA dominance by displacement of protein sequestered on triplet repeat RNA. Science 325, 336–339 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Furling, D. et al. Viral vector producing antisense RNA restores myotonic dystrophy myoblast functions. Gene Ther. 10, 795–802 (2003).

    Article  CAS  PubMed  Google Scholar 

  155. Mulders, S. A. M. et al. Triplet-repeat oligonucleotide-mediated reversal of RNA toxicity in myotonic dystrophy. Proc. Natl Acad. Sci. USA 106, 13915–13920 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  156. Majounie, E. et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol. 11, 323–330 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. DeJesus-Hernandez, M. et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72, 245–256 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Renton, A. E. et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72, 257–268 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Belzil, V. V., Gendron, T. F. & Petrucelli, L. RNA-mediated toxicity in neurodegenerative disease. Mol. Cell. Neurosci. 56, 406–419 (2013).

    Article  CAS  PubMed  Google Scholar 

  160. Gijselinck, I. et al. A C9orf72 promoter repeat expansion in a Flanders-Belgian cohort with disorders of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum: a gene identification study. Lancet Neurol. 11, 54–65 (2012).

    Article  CAS  PubMed  Google Scholar 

  161. Liu, E. Y. et al. C9orf72 hypermethylation protects against repeat expansion-associated pathology in ALS/FTD. Acta Neuropathol. 128, 525–541 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Xi, Z. et al. Hypermethylation of the CpG island near the G4C2 repeat in ALS with a C9orf72 expansion. Am. J. Hum. Genet. 92, 981–989 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Xi, Z. et al. The C9orf72 repeat expansion itself is methylated in ALS and FTLD patients. Acta Neuropathol. 129, 715–727 (2015).

    Article  CAS  PubMed  Google Scholar 

  164. Gendron, T. F., Belzil, V. V., Zhang, Y.-J. & Petrucelli, L. Mechanisms of toxicity in C9FTLD/ALS. Acta Neuropathol. 127, 359–376 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Ling, S.-C., Polymenidou, M. & Cleveland, D. W. Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron 79, 416–438 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Jiang, J. et al. Gain of toxicity from ALS/FTD-linked repeat expansions in C9ORF72 is alleviated by antisense oligonucleotides targeting GGGGCC-containing RNAs. Neuron 90, 535–550 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Zu, T. et al. Non-ATG-initiated translation directed by microsatellite expansions. Proc. Natl Acad. Sci. USA 108, 260–265 (2011).

    Article  PubMed  Google Scholar 

  168. Cleary, J. D. & Ranum, L. P. W. Repeat-associated non-ATG (RAN) translation in neurological disease. Hum. Mol. Genet. 22, R45–R51 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Freibaum, B. D. & Taylor, J. P. The role of dipeptide repeats in C9ORF72-related ALS-FTD. Front. Mol. Neurosci. 10, 35 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Lagier-Tourenne, C. et al. Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proc. Natl Acad. Sci. USA 110, E4530–E4539 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Donnelly, C. J. et al. RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron 80, 415–428 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Prior, T. W. & Finanger, E. in GeneReviews® (eds Adam, M. P. et al.) Ch. Spinal Muscular Atrophy (University of Washington, 2000).

    Google Scholar 

  173. Brzustowicz, L. M. et al. Genetic mapping of chronic childhood-onset spinal muscular atrophy to chromosome 5q1 1.2—13.3. Nature 344, 540–541 (1990).

    Article  CAS  PubMed  Google Scholar 

  174. Burghes, A. H. M. & Beattie, C. E. Spinal muscular atrophy: why do low levels of survival motor neuron protein make motor neurons sick? Nat. Rev. Neurosci. 10, 597–609 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Singh, N. K., Singh, N. N., Androphy, E. J. & Singh, R. N. Splicing of a critical exon of human Survival Motor Neuron is regulated by a unique silencer element located in the last intron. Mol. Cell. Biol. 26, 1333–1346 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Singh, N. N., Howell, M. D., Androphy, E. J. & Singh, R. N. How the discovery of ISS-N1 led to the first medical therapy for spinal muscular atrophy. Gene Ther. 24, 520–526 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Williams, J. H. et al. Oligonucleotide-mediated survival of motor neuron protein expression in CNS improves phenotype in a mouse model of spinal muscular atrophy. J. Neurosci. 29, 7633–7638 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Hua, Y. et al. Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes Dev. 24, 1634–1644 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Hua, Y. et al. Peripheral SMN restoration is essential for long-term rescue of a severe spinal muscular atrophy mouse model. Nature 478, 123–126 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Porensky, P. N. et al. A single administration of morpholino antisense oligomer rescues spinal muscular atrophy in mouse. Hum. Mol. Genet. 21, 1625–1638 (2012).

    Article  CAS  PubMed  Google Scholar 

  181. Zhou, H. et al. A novel morpholino oligomer targeting ISS-N1 improves rescue of severe spinal muscular atrophy transgenic mice. Hum. Gene Ther. 24, 331–342 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Boyer, J. G., Ferrier, A. & Kothary, R. More than a bystander: the contributions of intrinsic skeletal muscle defects in motor neuron diseases. Front. Physiol. 4, 356 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  183. Smith, L., Andersen, K. B., Hovgaard, L. & Jaroszewski, J. W. Rational selection of antisense oligonucleotide sequences. Eur. J. Pharm. Sci. 11, 191–198 (2000).

    Article  CAS  PubMed  Google Scholar 

  184. Echigoya, Y., Mouly, V., Garcia, L., Yokota, T. & Duddy, W. In silico screening based on predictive algorithms as a design tool for exon skipping oligonucleotides in Duchenne muscular dystrophy. PLoS ONE 10, e0120058 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Goyenvalle, A. et al. Functional correction in mouse models of muscular dystrophy using exon-skipping tricyclo-DNA oligomers. Nat. Med. 21, 270–275 (2015).

    Article  CAS  PubMed  Google Scholar 

  186. Iwamoto, N. et al. Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides. Nat. Biotechnol. 35, 845–851 (2017).

    Article  CAS  PubMed  Google Scholar 

  187. Alavijeh, M. S., Chishty, M., Qaiser, M. Z. & Palmer, A. M. Drug metabolism and pharmacokinetics, the blood-brain barrier, and central nervous system drug discovery. NeuroRx 2, 554–571 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  188. Bedi, D. et al. Targeted delivery of siRNA into breast cancer cells via phage fusion proteins. Mol. Pharm. 10, 551–559 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Falzarano, M. S., Passarelli, C. & Ferlini, A. Nanoparticle delivery of antisense oligonucleotides and their application in the exon skipping strategy for Duchenne muscular dystrophy. Nucleic Acid. Ther. 24, 87–100 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Lehto, T., Ezzat, K. & Wood, M. J. A. & EL Andaloussi, S. Peptides for nucleic acid delivery. Adv. Drug Deliv. Rev. 106, 172–182 (2016).

    Article  CAS  PubMed  Google Scholar 

  191. Ivanova, G. D. et al. PNA-peptide conjugates as intracellular gene control agents. Nucleic Acids Symp. Ser. 52, 31–32 (2008).

    Article  CAS  Google Scholar 

  192. Jearawiriyapaisarn, N. et al. Sustained dystrophin expression induced by peptide-conjugated morpholino oligomers in the muscles of mdx mice. Mol. Ther. 16, 1624–1629 (2008).

    Article  CAS  PubMed  Google Scholar 

  193. Wu, B. et al. Effective rescue of dystrophin improves cardiac function in dystrophin-deficient mice by a modified morpholino oligomer. Proc. Natl Acad. Sci. USA 105, 14814–14819 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  194. Yin, H. et al. Pip5 transduction peptides direct high efficiency oligonucleotide-mediated dystrophin exon skipping in heart and phenotypic correction in mdx mice. Mol. Ther. 19, 1295–1303 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Hammond, S. M. et al. Systemic peptide-mediated oligonucleotide therapy improves long-term survival in spinal muscular atrophy. Proc. Natl Acad. Sci. USA 113, 10962–10967 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Henry, S. P., Bolte, H., Auletta, C. & Kornbrust, D. J. Evaluation of the toxicity of ISIS 2302, a phosphorothioate oligonucleotide, in a four-week study in cynomolgus monkeys. Toxicology 120, 145–155 (1997).

    Article  CAS  PubMed  Google Scholar 

  197. Straarup, E. M. et al. Short locked nucleic acid antisense oligonucleotides potently reduce apolipoprotein B mRNA and serum cholesterol in mice and non-human primates. Nucleic Acids Res. 38, 7100–7111 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Iversen, P. L., Cornish, K. G., Iversen, L. J., Mata, J. E. & Bylund, D. B. Bolus intravenous injection of phosphorothioate oligonucleotides causes hypotension by acting as α1-adrenergic receptor antagonists. Toxicol. Appl. Pharmacol. 160, 289–296 (1999).

    Article  CAS  PubMed  Google Scholar 

  199. Lindow, M. et al. Assessing unintended hybridization-induced biological effects of oligonucleotides. Nat. Biotechnol. 30, 920–923 (2012).

    Article  CAS  PubMed  Google Scholar 

  200. Goemans, N. M. et al. Systemic administration of PRO051 in Duchenne's muscular dystrophy. N. Engl. J. Med. 364, 1513–1522 (2011).

    Article  CAS  PubMed  Google Scholar 

  201. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01128855 (2017).

  202. Voit, T. et al. Safety and efficacy of drisapersen for the treatment of Duchenne muscular dystrophy (DEMAND II): an exploratory, randomised, placebo-controlled phase 2 study. Lancet Neurol. 13, 987–996 (2014).

    Article  CAS  PubMed  Google Scholar 

  203. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01254019 (2017).

  204. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01480245 (2017).

  205. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01462292 (2017).

  206. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01803412 (2016).

  207. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02636686 (2016).

  208. Kinali, M., et al. Local restoration of dystrophin expression with the morpholino oligomer AVI-4658 in Duchenne muscular dystrophy: a single-blind, placebo-controlled, dose-escalation, proof-of-concept study. Lancet Neurol. 8, 918–928 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01540409 (2016).

  210. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02255552 (2017).

  211. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02286947 (2017).

  212. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02420379 (2017).

  213. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01037309 (2015).

  214. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01826474 (2015).

  215. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02500381 (2017).

  216. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01494701 (2017).

  217. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01703988 (2017).

  218. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01780246 (2017).

  219. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02052791 (2017).

  220. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02193074 (2017).

  221. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02292537 (2017).

  222. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02386553 (2017).

  223. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02462759 (2017).

  224. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02594124 (2017).

  225. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02865109 (2017).

  226. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01737398 (2016).

  227. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02519036 (2017).

  228. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02623699 (2017).

  229. Limmroth, V. et al. CD49d antisense drug ATL1102 reduces disease activity in patients with relapsing-remitting MS. Neurology 83, 1780–1788 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02312011 (2016).

Download references

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to the preparation of the article.

Corresponding author

Correspondence to Matthew J. A. Wood.

Ethics declarations

Competing interests

C.R. declares no competing interests. M.J.A.W., through the University of Oxford, has filed patents on peptide-based methods for antisense oligonucleotide delivery.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rinaldi, C., Wood, M. Antisense oligonucleotides: the next frontier for treatment of neurological disorders. Nat Rev Neurol 14, 9–21 (2018). https://doi.org/10.1038/nrneurol.2017.148

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrneurol.2017.148

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing