Yonsei Med J.  2020 Apr;61(4):273-283. 10.3349/ymj.2020.61.4.273.

Nusinersen as a Therapeutic Agent for Spinal Muscular Atrophy

Affiliations
  • 1Department of Function, ShiJiaZhuang Traditional Chinese Medical Hospital, ShiJiaZhuang, HeBei, China. liqingliyang@126.com

Abstract

The reduction of survival motor neuron (SMN) protein causes spinal muscular atrophy (SMA), an autosomal recessive neuromuscular disease. Nusinersen is an antisense oligonucleotide, approved by the FDA, which specifically binds to the repressor within SMN2 exon 7 to enhance exon 7 inclusion and augment production of functional SMN protein. Nusinersen is the first new oligonucleotide-based drug targeting the central nervous system for the treatment of SMA. This review of nusinersen will discuss its action mechanism, cellular uptake, trafficking mechanisms, and administration approaches to cross the blood-brain barrier. Furthermore, nusinersen clinical trials will be assessed in terms of pharmacokinetics, tolerability and safety, the clinical outcomes of multiple intrathecal doses, and a discussion on the primary and secondary endpoints.

Keyword

blood-brain barrier; cellular uptake; exon splicing; phosphonothioate antisense oligonucleotides; survival of motor neuron gene; Nusinersen

MeSH Terms

Blood-Brain Barrier
Central Nervous System
Drug Delivery Systems
Exons
Motor Neurons
Muscular Atrophy, Spinal*
Neuromuscular Diseases
Pharmacokinetics

Figure

  • Fig. 1 Oligonucleotide uptake and trafficking in cells. It has been proposed that phosphonothioate antisense oligonucleotides (PS-ASOs) enter cells via two pathways: productive pathway and non-productive pathway. Productive pathway guides PS-ASOs to access their targets (yellow outlines), and non-productive pathways sequester PS-ASOs in saturable sinks (blue outlines). Uptake and internalization of PS-ASOs can be directed by cell-surface proteins (including receptors) through caveolin- or clathrin-dependent pathways or other pathways of endocytosis. The endocytosis vesicle bud from the membrane and the scission of the plasma membrane is catalyzed by GTPase dynamin. Freed cargo-containing vesicles with PS-ASOs fuse with early endosomes (EE) by being transported along the cytoskeletal structures of tubulin or actin. Internalized ASOs can be transported from EE to late endosomes (LE) and to lysosomes. PS-ASOs predominately accumulate in punctate structures in EE, LE, lysosomes (possibly equal to phosphonothioate bodies of cytoplasm), and Golgi-58K-related vesicles, in which non-productive and productive pathways of uptake may diverge and the majority of oligonucleotides are likely reserved in non-productive pathway. A small number of PS-ASOs can be released from endosomal organelles and escape into productive pathway, involving entry into the nucleus and cytosol to act on target DNA. Entering into the nucleus is likely via the RAN-mediated pathway. PS-ASOs may be linked to paraspeckle proteins in the nucleus and shape functional paraspeckle-like structures.


Reference

1. Hua Y, Sahashi K, Hung G, Rigo F, Passini MA, Bennett CF, et al. Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes Dev. 2010; 24:1634–1644.
Article
2. Singh NK, Singh NN, Androphy EJ, Singh RN. 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. 2006; 26:1333–1346.
Article
3. Wan L, Battle DJ, Yong J, Gubitz AK, Kolb SJ, Wang J, et al. The survival of motor neurons protein determines the capacity for snRNP assembly: biochemical deficiency in spinal muscular atrophy. Mol Cell Biol. 2005; 25:5543–5551.
Article
4. Hua Y, Vickers TA, Baker BF, Bennett CF, Krainer AR. Enhancement of SMN2 exon 7 inclusion by antisense oligonucleotides targeting the exon. PLoS Biol. 2007; 5:e73.
Article
5. Kashima T, Manley JL. A negative element in SMN2 exon 7 inhibits splicing in spinal muscular atrophy. Nat Genet. 2003; 34:460–463.
Article
6. Cartegni L, Krainer AR. Disruption of an SF2/ASF-dependent exonic splicing enhancer in SMN2 causes spinal muscular atrophy in the absence of SMN1. Nat Genet. 2002; 30:377–384.
Article
7. Lunn MR, Wang CH. Spinal muscular atrophy. Lancet. 2008; 371:2120–2133.
Article
8. Foust KD, Wang X, McGovern VL, Braun L, Bevan AK, Haidet AM, et al. Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nat Biotechnol. 2010; 28:271–274.
Article
9. Coady TH, Shababi M, Tullis GE, Lorson CL. Restoration of SMN function: delivery of a trans-splicing RNA re-directs SMN2 premRNA splicing. Mol Ther. 2007; 15:1471–1478.
Article
10. Madocsai C, Lim SR, Geib T, Lam BJ, Hertel KJ. Correction of SMN2 Pre-mRNA splicing by antisense U7 small nuclear RNAs. Mol Ther. 2005; 12:1013–1022.
Article
11. Khorkova O, Wahlestedt C. Oligonucleotide therapies for disorders of the nervous system. Nat Biotechnol. 2017; 35:249–263.
Article
12. Farr SA, Erickson MA, Niehoff ML, Banks WA, Morley JE. Central and peripheral administration of antisense oligonucleotide targeting amyloid-β protein precursor improves learning and memory and reduces neuroinflammatory cytokines in Tg2576 (AβPPswe) mice. J Alzheimers Dis. 2014; 40:1005–1016.
Article
13. Juliano RL. The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 2016; 44:6518–6548.
Article
14. Eckstein F. Nucleoside phosphorothioates. J Am Chem Soc. 1970; 92:4718–4723.
Article
15. Bobst AM, Rottman F, Cerutti PA. Effect of the methylation of the 2′-hydroxyl groups in polyadenylic acid on its structure in weakly acidic and neutral solutions and on its capability to form ordered complexes with polyuridylic acid. J Mol Biol. 1969; 46:221–234.
Article
16. Evers MM, Toonen LJ, van Roon-Mom WM. Antisense oligonucleotides in therapy for neurodegenerative disorders. Adv Drug Deliv Rev. 2015; 87:90–103.
Article
17. Hua Y, Vickers TA, Okunola HL, Bennett CF, Krainer AR. Antisense masking of an hnRNP A1/A2 intronic splicing silencer corrects SMN2 splicing in transgenic mice. Am J Hum Genet. 2008; 82:834–848.
Article
18. Rigo F, Hua Y, Krainer AR, Bennett CF. Antisense-based therapy for the treatment of spinal muscular atrophy. J Cell Biol. 2012; 199:21–25.
Article
19. Juliano RL, Ming X, Nakagawa O. Cellular uptake and intracellular trafficking of antisense and siRNA oligonucleotides. Bioconjug Chem. 2012; 23:147–157.
Article
20. Koller E, Vincent TM, Chappell A, De S, Manoharan M, Bennett CF. Mechanisms of single-stranded phosphorothioate modified antisense oligonucleotide accumulation in hepatocytes. Nucleic Acids Res. 2011; 39:4795–4807.
Article
21. Wagenaar TR, Tolstykh T, Shi C, Jiang L, Zhang J, Li Z, 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. 2015; 43:1204–1215.
Article
22. Liang XH, Shen W, Sun H, Kinberger GA, Prakash TP, Nichols JG, et al. Hsp90 protein interacts with phosphorothioate oligonucleotides containing hydrophobic 2′-modifications and enhances antisense activity. Nucleic Acids Res. 2016; 44:3892–3907.
Article
23. Shemesh CS, Yu RZ, Gaus HJ, Seth PP, Swayze EE, Bennett FC, et al. Pharmacokinetic and pharmacodynamic investigations of ION-353382, a model antisense oligonucleotide: using alpha-2-macroglobulin and murinoglobulin double-knockout mice. Nucleic Acid Ther. 2016; 26:223–235.
Article
24. Shen W, Liang XH, Crooke ST. Phosphorothioate oligonucleotides can displace NEAT1 RNA and form nuclear paraspeckle-like structures. Nucleic Acids Res. 2014; 42:8648–8662.
Article
25. Liang XH, Shen W, Sun H, Prakash TP, Crooke ST. TCP1 complex proteins interact with phosphorothioate oligonucleotides and can co-localize in oligonucleotide-induced nuclear bodies in mammalian cells. Nucleic Acids Res. 2014; 42:7819–7832.
Article
26. Liang XH, Sun H, Shen W, Crooke ST. Identification and characterization of intracellular proteins that bind oligonucleotides with phosphorothioate linkages. Nucleic Acids Res. 2015; 43:2927–2945.
Article
27. Ezzat K, Helmfors H, Tudoran O, Juks C, Lindberg S, Padari K, et al. Scavenger receptor-mediated uptake of cell-penetrating peptide nanocomplexes with oligonucleotides. FASEB J. 2012; 26:1172–1180.
Article
28. Ugarte-Uribe B, Pérez-Rentero S, Lucas R, Aviñó A, Reina JJ, Alkorta I, et al. Synthesis, cell-surface binding, and cellular uptake of fluorescently labeled glucose-DNA conjugates with different carbohydrate presentation. Bioconjug Chem. 2010; 21:1280–1287.
Article
29. Cheng CJ, Saltzman WM. Enhanced siRNA delivery into cells by exploiting the synergy between targeting ligands and cell-penetrating peptides. Biomaterials. 2011; 32:6194–6203.
Article
30. Alam MR, Dixit V, Kang H, Li ZB, Chen X, Trejo J, et al. Intracellular delivery of an anionic antisense oligonucleotide via receptor-mediated endocytosis. Nucleic Acids Res. 2008; 36:2764–2776.
Article
31. Juliano RL, Carver K, Cao C, Ming X. Receptors, endocytosis, and trafficking: the biological basis of targeted delivery of antisense and siRNA oligonucleotides. J Drug Target. 2013; 21:27–43.
Article
32. Juliano RL, Ming X, Carver K, Laing B. Cellular uptake and intracellular trafficking of oligonucleotides: implications for oligonucleotide pharmacology. Nucleic Acid Ther. 2014; 24:101–113.
Article
33. Sahay G, Querbes W, Alabi C, Eltoukhy A, Sarkar S, Zurenko C, et al. Efficiency of siRNA delivery by lipid nanoparticles is limited by endocytic recycling. Nat Biotechnol. 2013; 31:653–658.
Article
34. Ming X, Alam MR, Fisher M, Yan Y, Chen X, Juliano RL. Intracellular delivery of an antisense oligonucleotide via endocytosis of a G protein-coupled receptor. Nucleic Acids Res. 2010; 38:6567–6576.
Article
35. Miller CM, Donner AJ, Blank EE, Egger AW, Kellar BM, Østergaard ME, et al. Stabilin-1 and Stabilin-2 are specific receptors for the cellular internalization of phosphorothioate-modified antisense oligonucleotides (ASOs) in the liver. Nucleic Acids Res. 2016; 44:2782–2794.
Article
36. Kortylewski M, Swiderski P, Herrmann A, Wang L, Kowolik C, Kujawski M, et al. In vivo delivery of siRNA to immune cells by conjugation to a TLR9 agonist enhances antitumor immune responses. Nat Biotechnol. 2009; 27:925–932.
Article
37. Shi F, Sottile J. Caveolin-1-dependent beta1 integrin endocytosis is a critical regulator of fibronectin turnover. J Cell Sci. 2008; 121(Pt 14):2360–2371.
Article
38. Juliano RL, Carver K. Cellular uptake and intracellular trafficking of oligonucleotides. Adv Drug Deliv Rev. 2015; 87:35–45.
Article
39. Reyes-Reyes EM, Teng Y, Bates PJ. A new paradigm for aptamer therapeutic AS1411 action: uptake by macropinocytosis and its stimulation by a nucleolin-dependent mechanism. Cancer Res. 2010; 70:8617–8629.
Article
40. Ezzat K, Aoki Y, Koo T, McClorey G, Benner L, Coenen-Stass A, et al. Self-assembly into nanoparticles is essential for receptor mediated uptake of therapeutic antisense oligonucleotides. Nano Lett. 2015; 15:4364–4373.
Article
41. Kotula JW, Pratico ED, Ming X, Nakagawa O, Juliano RL, Sullenger BA. Aptamer-mediated delivery of splice-switching oligonucleotides to the nuclei of cancer cells. Nucleic Acid Ther. 2012; 22:187–195.
Article
42. Wang S, Sun H, Tanowitz M, Liang XH, Crooke ST. Annexin A2 facilitates endocytic trafficking of antisense oligonucleotides. Nucleic Acids Res. 2016; 44:7314–7330.
Article
43. Prakash TP, Graham MJ, Yu J, Carty R, Low A, Chappell A, et al. Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice. Nucleic Acids Res. 2014; 42:8796–8807.
Article
44. Vickers TA, Crooke ST. Development of a quantitative BRET affinity assay for nucleic acid-protein interactions. PLoS One. 2016; 11:e0161930.
Article
45. Castanotto D, Lin M, Kowolik C, Wang L, Ren XQ, Soifer HS, et al. A cytoplasmic pathway for gapmer antisense oligonucleotide-mediated gene silencing in mammalian cells. Nucleic Acids Res. 2015; 43:9350–9361.
Article
46. Lorenz P, Baker BF, Bennett CF, Spector DL. Phosphorothioate antisense oligonucleotides induce the formation of nuclear bodies. Mol Biol Cell. 1998; 9:1007–1023.
Article
47. Kubo T, Zhelev Z, Bakalova R, Ohba H, Doi K, Fujii M. Controlled intracellular localization and enhanced antisense effect of oligonucleotides by chemical conjugation. Org Biomol Chem. 2005; 3:3257–3259.
Article
48. Marcusson EG, Bhat B, Manoharan M, Bennett CF, Dean NM. Phosphorothioate oligodeoxyribonucleotides dissociate from cationic lipids before entering the nucleus. Nucleic Acids Res. 1998; 26:2016–2023.
Article
49. Zelphati O, Szoka FC Jr. Mechanism of oligonucleotide release from cationic liposomes. Proc Natl Acad Sci U S A. 1996; 93:11493–11498.
Article
50. Naganuma T, Hirose T. Paraspeckle formation during the biogenesis of long non-coding RNAs. RNA Biol. 2013; 10:456–461.
Article
51. Lorenz P, Misteli T, Baker BF, Bennett CF, Spector DL. Nucleocytoplasmic shuttling: a novel in vivo property of antisense phosphorothioate oligodeoxynucleotides. Nucleic Acids Res. 2000; 28:582–592.
Article
52. Doherty GJ, McMahon HT. Mechanisms of endocytosis. Annu Rev Biochem. 2009; 78:857–902.
Article
53. Stein CA, Hansen JB, Lai J, Wu S, Voskresenskiy A, Høg A, et al. Efficient gene silencing by delivery of locked nucleic acid antisense oligonucleotides, unassisted by transfection reagents. Nucleic Acids Res. 2010; 38:e3.
Article
54. Torres AG, Threlfall RN, Gait MJ. Potent and sustained cellular inhibition of miR-122 by lysine-derivatized peptide nucleic acids (PNA) and phosphorothioate locked nucleic acid (LNA)/2′-O-methyl (OMe) mixmer anti-miRs in the absence of transfection agents. Artif DNA PNA XNA. 2011; 2:71–78.
Article
55. Smith RA, Miller TM, Yamanaka K, Monia BP, Condon TP, Hung G, et al. Antisense oligonucleotide therapy for neurodegenerative disease. J Clin Invest. 2006; 116:2290–2296.
Article
56. Wahlestedt C, Golanov E, Yamamoto S, Yee F, Ericson H, Yoo H, et al. Antisense oligodeoxynucleotides to NMDA-R1 receptor channel protect cortical neurons from excitotoxicity and reduce focal ischaemic infarctions. Nature. 1993; 363:260–263.
Article
57. Wahlestedt C, Pich EM, Koob GF, Yee F, Heilig M. Modulation of anxiety and neuropeptide Y-Y1 receptors by antisense oligodeoxynucleotides. Science. 1993; 259:528–531.
Article
58. Wahlestedt C. Antisense oligonucleotide strategies in neuropharmacology. Trends Pharmacol Sci. 1994; 15:42–46.
Article
59. Standifer KM, Chien CC, Wahlestedt C, Brown GP, Pasternak GW. Selective loss of delta opioid analgesia and binding by antisense oligodeoxynucleotides to a delta opioid receptor. Neuron. 1994; 12:805–810.
Article
60. Pechmann A, Langer T, Wider S, Kirschner J. Single-center experience with intrathecal administration of nusinersen in children with spinal muscular atrophy type 1. Eur J Paediatr Neurol. 2018; 22:122–127.
Article
61. Zanetta C, Nizzardo M, Simone C, Monguzzi E, Bresolin N, Comi GP, et al. Molecular therapeutic strategies for spinal muscular atrophies: current and future clinical trials. Clin Ther. 2014; 36:128–140.
Article
62. Passini MA, Bu J, Richards AM, Kinnecom C, Sardi SP, Stanek LM, et al. Antisense oligonucleotides delivered to the mouse CNS ameliorate symptoms of severe spinal muscular atrophy. Sci Transl Med. 2011; 3:72ra18.
Article
63. Geary RS. Antisense oligonucleotide pharmacokinetics and metabolism. Expert Opin Drug Metab Toxicol. 2009; 5:381–391.
Article
64. Rigo F, Chun SJ, Norris DA, Hung G, Lee S, Matson J, 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. 2014; 350:46–55.
Article
65. Heemskerk H, de Winter C, van Kuik P, Heuvelmans N, Sabatelli P, Rimessi P, et al. Preclinical PK and PD studies on 2′-O-methyl-phosphorothioate RNA antisense oligonucleotides in the mdx mouse model. Mol Ther. 2010; 18:1210–1217.
Article
66. Verhaart IE, Tanganyika-de Winter CL, Karnaoukh TG, Kolfschoten IG, de Kimpe SJ, van Deutekom JC, et al. Dose-dependent pharmacokinetic profiles of 2′-O-methyl phosphorothioate antisense oligonucleotidesin mdx mice. Nucleic Acid Ther. 2013; 23:228–237.
Article
67. Kordasiewicz HB, Stanek LM, Wancewicz EV, Mazur C, McAlonis MM, Pytel KA, et al. Sustained therapeutic reversal of Huntington's disease by transient repression of huntingtin synthesis. Neuron. 2012; 74:1031–1044.
Article
68. Geary RS, Norris D, Yu R, Bennett CF. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv Drug Deliv Rev. 2015; 87:46–51.
Article
69. Chiriboga CA, Swoboda KJ, Darras BT, Iannaccone ST, Montes J, De Vivo DC, et al. Results from a phase 1 study of nusinersen (ISIS-SMN(Rx)) in children with spinal muscular atrophy. Neurology. 2016; 86:890–897.
Article
70. Haché M, Swoboda KJ, Sethna N, Farrow-Gillespie A, Khandji , Xia S, et al. Intrathecal injections in children with spinal muscular atrophy: nusinersen clinical trial experience. J Child Neurol. 2016; 31:899–906.
Article
71. Engelhardt JA. Comparative renal toxicopathology of antisense oligonucleotides. Nucleic Acid Ther. 2016; 26:199–209.
Article
72. Matsuzawa J, Matsui M, Konishi T, Noguchi K, Gur RC, Bilker W, et al. Age-related volumetric changes of brain gray and white matter in healthy infants and children. Cereb Cortex. 2001; 11:335–342.
Article
73. Finkel RS, Chiriboga CA, Vajsar J, Day JW, Montes J, De Vivo DC, et al. Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study. Lancet. 2016; 388:3017–3026.
Article
74. Frisone MF, Mercuri E, Laroche S, Foglia C, Maalouf EF, Haataja L, et al. Prognostic value of the neurologic optimality score at 9 and 18 months in preterm infants born before 31 weeks' gestation. J Pediatr. 2002; 140:57–60.
Article
75. Glanzman AM, Mazzone E, Main M, Pelliccioni M, Wood J, Swoboda KJ, et al. The Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP INTEND): test development and reliability. Neuromuscul Disord. 2010; 20:155–161.
Article
76. Yu RZ, Baker B, Chappell A, Geary RS, Cheung E, Levin AA. Development of an ultrasensitive noncompetitive hybridization-ligation enzyme-linked immunosorbent assay for the determination of phosphorothioate oligodeoxynucleotide in plasma. Anal Biochem. 2002; 304:19–25.
Article
77. Finkel RS, Mercuri E, Darras BT, Connolly AM, Kuntz NL, Kirschner J, et al. Nusinersen versus sham control in infantile-onset spinal muscular atrophy. N Engl J Med. 2017; 377:1723–1732.
Article
78. Mercuri E, Darras BT, Chiriboga CA, Day JW, Campbell C, Connolly AM, et al. Nusinersen versus sham control in later-onset spinal muscular atrophy. N Engl J Med. 2018; 378:625–635.
Article
79. Glanzman AM, O'Hagen JM, McDermott MP, Martens WB, Flickinger J, Riley S, et al. Validation of the Expanded Hammersmith Functional Motor Scale in spinal muscular atrophy type II and III. J Child Neurol. 2011; 26:1499–1507.
Article
80. O'Hagen JM, Glanzman AM, McDermott MP, Ryan PA, Flickinger J, Quigley J, et al. An expanded version of the Hammersmith Functional Motor Scale for SMA II and III patients. Neuromuscul Disord. 2007; 17:693–697.
81. Ebinger F, Kosel C, Pietz J, Rating D. Headache and backache after lumbar puncture in children and adolescents: a prospective study. Pediatrics. 2004; 113:1588–1592.
Article
82. Morgenlander JC. Lumbar puncture and CSF examination. Answers to three commonly asked questions. Postgrad Med. 1994; 95:125–128.
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr