Recent advances in genetic testing have accelerated the genetic diagnosis of rare diseases; however, the development of therapeutics has progressed comparatively slowly [1]. Currently only 5% of rare genetic disorders have approved treatments and 15% are undergoing clinical trials [2]. Considering that 80% of rare genetic disorders have neurological involvement, early treatment is crucial for most of these patients, given the rapidly progressive and fatal nature of the diseases, especially in pediatric patients with a developmental time frame [3].
Monogenic disorders account for 80% of rare genetic disorders [4]. Thus, gene-based treatment has become more of an interest in the clinical and research fields. Gene-based therapies involve both DNA- and RNA-directed oligonucleotide treatments. DNA-directed approaches include gene transfer via viral or non-viral vectors and gene-editing techniques such as CRISPR-Cas9, base editing, and prime editing. These methods enable precise genetic modifications and potentially correct disease-causing mutations at the DNA level. However, gene transfer requires a viral vector that can activate the immune system. Gene expression depends on the stability of the episomal forms. These effects may wane, particularly in rapidly dividing tissues. In contrast, RNA-directed therapies do not require a viral vector and messenger RNA (mRNA) is continuously replenished, ensuring stability and sustained expression. RNA-targeting drugs, including antisense oligonucleotides (ASOs), small interfering RNA (siRNA), mRNA therapy, and RNA editing, also exhibit high selectivity owing to their nucleotide sequence specificity, effectively targeting the intended sites while maintaining stability.
In this review, we discuss the basic mechanisms and latest clinical applications of ASOs in rare neurogenetic disorders. ASOs have gained considerable attention due to the clinical success of nusinersen (Spinraza), an ASO that modifies pre-mRNA splicing of SMN2 to promote increased production of full-length SMN protein [5]. This breakthrough has encouraged further development of ASOs for rare genetic disorders. Furthermore, the first personalized ASO was proven successful in a female child with Batten disease, which opened a new paradigm for n-of-1 treatment in the world of oligonucleotide therapeutics. The significance of ASOs was underscored by Kim et al. [6], who reported that ASOs show therapeutic potential in approximately 9-15% of rare genetic diseases, highlighting their growing impact in precision medicine regarding rare neurogenetic disorders.
ASOs typically comprise 12-25 nucleotides and attach to complementary RNA sequences via Watson–Crick base pairing. ASOs can target both protein-coding and non-coding RNA to modulate protein expression. Their small size (4-10 kDa), single-stranded structure, and hydrophilic properties enable spontaneous cellular uptake without the need for carriers, similar to that of small-molecule drugs. ASOs can penetrate the brain, spinal cord, and retina without chemical modifications; however, modifications can enhance their delivery to the liver or muscles. Among gene-based therapies, ASOs are notable for their minimal off-target and side effects combined with the benefit of chemically stable modifications [7].
The mechanisms of action primarily involve RNA cleavage and blockage, shown in detail in Fig. 1 [8,9]. RNA cleavage is facilitated by the recruitment of RNase H, an enzyme that specifically degrades the RNA strand of a DNA-RNA hybrid. ASOs, which are designed as gapmers, contain a central DNA gap flanked by chemically modified RNA segments that enhance their stability and binding affinity. The DNA region of the gapmer hybridizes with the target RNA, forming a duplex that activates RNase H, leading to the selective degradation of the target RNA. This mechanism may be used to knock down target transcripts and downregulate mRNA expression (Fig. 1A).
In contrast, RNA blockage occurs through steric hindrance or modulation of RNA splicing. ASOs can interact with pre-mRNAs to influence splicing events, including the skipping of exons, to potentially correct splicing defects or alter the production of specific protein isoforms (Figs. 1B and C).
Furthermore, ASOs could be used to increase the production of proteins from the wild-type (WT) transcript by upregulating the WT allele via targeted augmentation of nuclear gene output (TANGO), which augments the targeted area of the WT allele to induce greater production of functional proteins [10]. An ASO can be created to skip poison exons, target antisense transcripts (ASTs), and untranslated regions, including upstream open reading frames (uORFs) (Fig. 1D). Poison exons are conserved sequences that, when included in mRNA, lead to the early termination of translation. The use of ASOs to skip these exons boosts the generation of functional transcripts, thereby increasing the protein expression. ASTs are non-coding RNAs that regulate gene expression via RNA interference or masking. ASOs may inhibit the expression of AST to increase the protein expression. uORFs are defined as translation sites found within the 5’ untranslated region of mRNAs, positioned before the main protein-coding sequence. ASOs can both enhance or inhibit protein expression by blocking or skipping exons containing uORFs [11].
Recent advancements in the chemical modification of ASOs have resulted in three generations. The first generation consists of a phosphorothioate (PS) backbone, which involves substituting the non-bridging oxygen in the phosphate backbone with sulfur. Second-generation modifications include 2‘-O-modifications such as 2‘-O-methoxyethyl (2‘-MOE), 2‘-O-methyl (2‘-OMe), and 2‘-fluoro (2‘-F), which improve RNA affinity and metabolic stability. Third-generation modifications include the locked nucleic acid (LNA) method. LNA modifications lock the ribose sugar in a specific conformation, which significantly enhances binding affinity and stability. Furthermore, emerging strategies include conjugating ASOs with targeting ligands such as GalNAc, which enhance tissue delivery and minimize side effects [12,13]. These modifications enhance ASO stability, binding affinity, and cellular uptake, while reducing nuclease susceptibility, thereby prolonging its half-life in the bloodstream and tissues [9,14]. Therefore, ASOs achieve high specificity and efficacy in targeting and modulating gene expression, making them promising therapeutic options for a wide range of genetic disorders.
As of April 2024, 19 oligonucleotide therapies have been approved by the New Drug Application (NDA) of the United States Food and Drug Administration (US FDA) and the European Medicines Agency (EMA) for the treatment of rare genetic disorders [12,15]. Eight of the 19 oligonucleotide therapies were designated as ASOs for rare neurogenetic disorders (Table 1). Furthermore, several n-of-1 personalized ASOs have been granted approval under Investigational New Drug (IND) status (Table 2).
Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder, the majority of which is caused by a homozygous deletion of the survival motor neuron 1 (
Duchenne muscular dystrophy (DMD), an X-linked recessive disorder caused by mutations in the dystrophin gene (Xp21), results in progressive skeletal muscle degeneration and weakness. Approximately 65% of patients have exonic deletions, 5-10% have exonic duplications, and the remainder are mainly nonsense and indel mutations. Four ASOs, using phosphorodiamidate morpholino oligomers (PMO) to induce exon skipping, have been approved: Eteplirsen (Exondys 51, exon 51, FDA 2016) [20], Golodirsen (Vyondys 53, exon 53, FDA 2019) [21], Viltolarsen (Viltepso, exon 53, FDA 2020) [22], and Casimersen (Amondys 45, exon 45, FDA 2021) [23]. The ASOs are administered weekly via intravenous injections. All four ASOs have shown modest improvements by increasing dystrophin levels and slowing disease progression. Clinical outcomes vary with a limited impact on long-term ambulation and mixed results regarding the ability to preserve motor function in patients [24,25]. Observational studies, such as the EVOLVE trial, have highlighted favorable safety profiles; however, ongoing research is needed to confirm their long-term efficacy and functional benefits [26].
Hereditary transthyretin-mediated amyloidosis (hATTR), an autosomal dominant disorder resulting from mutations in the
Amyotrophic lateral sclerosis (ALS) selectively affects the motor neurons and progressively destroys the brain, brainstem, and spinal cord. Approximately 5-10% of ALS cases have a genetic etiology, with approximately 20% linked to mutations in
The advent of personalized ASOs has provided a breakthrough for patients with neurogenetic disorders who lack effective treatment options. A notable case involved a child with Batten disease who received a splice-modulating ASO, resulting in marked clinical improvement and demonstrating the potential for individualized genetic therapies. This case set a precedent and inspired further global initiatives. Leading efforts in personalized ASO development include programs such as N=1 Collaborative, n-Lorem, Dutch Center for RNA Therapeutics (DCRT), and 1 Mutation 1 Medicine (1M1M). These organizations have pioneered customized therapies for ultra-rare conditions, with several impactful cases documented. Anecdotally, more than 20 projects involving patient-customized ASOs are ongoing internationally, some of which are listed in Table 2 [6,33-41]. This progress highlights a promising shift toward precision medicine and the transformation of care for patients with otherwise untreatable genetic diseases.
Milasen was the first personalized ASO therapy targeting a specific mutation in Mila, a six-year-old with Batten disease [33]. Batten disease leads to vision loss, seizures, limb paralysis, and death. Mila’s condition was caused by a SINE-VNTR-Alu (SVA) retrotransposon insertion in the
Following the success of Milasen, the second personalized ASO therapy, Atipeksen, was developed for Ipek, who was diagnosed with ataxia telangiectasia (AT) at age 1 after newborn screening revealed low T-cell receptor excision circles [6]. AT, caused by mutations in the
Following the two splice-modulating n-of-1 ASOs, the first knockdown gapmer ASO was successful in a patient with
There are many emerging ASOs in the process of moving from the laboratory bench to the bedside, offering hope for patients with previously untreatable neurogenetic disorders. Table 3 summarizes the ASOs currently being administered to patients and those undergoing clinical trials. Before reaching this stage, these ASOs had already demonstrated therapeutic potential in patient-derived cell lines and
Dravet syndrome is characterized by seizures during early childhood and is often accompanied by developmental disorders [44]. More than 80% of cases are associated with
Mutations in
Angelman syndrome manifests with severe developmental delay, epilepsy, and other symptoms. This is due to the loss of function of the maternally inherited
ASOs have redefined the landscape of rare neurogenetic disorders by delivering targeted therapies that address their root causes at the molecular level. Their impact is exemplified by breakthrough treatments, such as nusinersen and personalized ASOs, which show how precision interventions can alter disease trajectories and enhance the quality of life. The integration of evolving diagnostics with precision therapeutics will not only enable early detection, but also allow timely, individualized treatment to prevent irreversible progression. As advancements in delivery systems and chemical modifications continue to enhance ASO efficacy, these therapies are poised to become a foundational pillar of precision medicine, transforming the care for patients with previously untreatable genetic diseases.
We acknowledge the Medical Illustration & Design (MID) team, a member of the Medical Research Support Services of Yonsei University College of Medicine, for providing excellent support with the medical illustration.
This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: RS-2024-00405260 and RS-2024-00439002).
Conception and design: HJS, HCK. Acquisition of data: HJS, AK, JYO. Analysis and interpretation of data: HJS, AK, JYO. Drafting the article: HJS. Critical revision of the article: AK, HCK. Final approval of the version to be published: HCK.
FDA-approved antisense oligonucleotides for rare neurogenetic disorders (n=8)
Target gene | Disease | ASO/mechanism/chemical modification | Route/interval of administration | FDA-approval |
---|---|---|---|---|
Spinal muscular atrophy | Nusinersen (Spinraza), splice modulating, 18-mer 2’-MOE-modified ASO | Intrathecal, every 4 months | 2016 | |
Duchenne muscular dystrophy | Eteplirsen (Exondys 51), exon skipping, PMO | Intravenous, weekly | 2016 | |
Duchenne muscular dystrophy | Golodirsen (Vyondys), exon skipping, PMO | Intravenous, weekly | 2019 | |
Duchenne muscular dystrophy | Viltolarsen (Viltepso), exon skipping, PMO | Intravenous, weekly | 2020 | |
Duchenne muscular dystrophy | Casimersen (Amondys 45), exon skipping, PMO | Intravenous, weekly | 2021 | |
Hereditary transthyretin-mediated amyloidosis | Inotersen (Tegsedi), a knockdown 2’-MOE gapmer ASO, PS linkage | Subcutaneous, weekly | 2018 | |
Hereditary transthyretin-mediated amyloidosis | Eplontersen (Wainua), a gapmer ASO, PO linkage and GalNAc ligand conjugation | Subcutaneous, weekly | 2023 | |
Amyotrophic lateral sclerosis | Tofersen (Qalsody), an 2’-MOE gapmer | Intrathecal, monthly | 2023 |
FDA, Food and Drug Administration; ASO, antisense oligonucleotide; 2’-MOE, 2’-O-methoxyethyl; PMO, phosphorodiamidate morpholino oligomer; PS, phosphorothioate; PO, phosphodiester.
Personalized antisense oligonucleotides for rare neurogenetic disorders
Target gene | Disease | ASO/mechanism | Method of administration | Study data | Principal investigator/sponsor |
---|---|---|---|---|---|
Batten disease | Splice modulating, 2’-MOE-modified ASO | Intrathecal | n=1, decreased seizure frequency | Boston Children’s Hospital [33] | |
Ataxia telengiectasia | Splice modulating, 2’-MOE-modified ASO | Intrathecal | n=2, milder neurological impairment | Boston Children’s Hospital [6,34,35] | |
DEE | Knockdown, gapmer ASO | Intrathecal | n=2, decreased seizure frequency, paused due to ventriculomegaly | Boston Children’s Hospital [35,36] | |
Neurological disorder | Knockdown, gapmer ASO | Intrathecal, every 3 months | n=2, decreased seizure frequency, improvement in development | Boston Children’s Hospital, n-Lorem Foundation [37] | |
PCARP | Splice-modulating | Intravitreal, every 3 months | n=1, unknown | Boston Children’s Hospital, n-Lorem Foundation [38] | |
DEE | Knockdown, gapmer ASO | Intrathecal | n=1, decreased seizure frequency | Rady Children’s Hospital, UCSD, n-Lorem Foundation, NCT06314490 [38] | |
Leukodystrophy | Knockdown, gapmer ASO | Unknown | n=1, unknown | Massachusetts General Hospital, n-Lorem Foundation, NCT06369974 | |
Batten disease | Splice-modulating | Unknown | n=2 (twins), positive outcome | University of North Carolina, Vanguard Clinical Inc. [39,40] | |
ALS | Unknown | Unknown | n=1, unknown | Mayo Clinic, n-Lorem Foundation, NCT06392126 | |
Transportin-associated disorder | Knockdown, gapmer ASO, locked nucleic acid conjugation | Intrathecal, every 3 months | n=1, improved development, decreased seizure frequency | Rady Children’s Hospital, UCSD, Creyon Bio, Inc. [35,41] |
ASO, antisense oligonucleotide; 2’-MOE, 2’-O-methoxyethyl; DEE, developmental and epileptic encephalopathy; PCARP, posterior column ataxia and retinitis pigmentosa; UCSD, University of California San Diego; ALS, amyotrophic lateral aclerosis.
Emerging antisense oligonucleotides for rare neurogenetic disorders undergoing clinical trials
Trial | Target gene | Disease | ASO/mechanism | Route/interval of administration | Study status | Sponsor |
---|---|---|---|---|---|---|
NCT04740476 | Dravet syndrome | STK-001, TANGO | Intrathecal | n=60 (enrolling), age ≥30 months, phase 2 | Stoke Therapeutics | |
NCT05737784 | DEE, gain of function | Elsunersen, knockdown gapmer | Intrathecal, every 4 weeks | n=60 (enrolling), age 2-18 years, phase 2 | Praxis Precision Medicines | |
NCT04259281 | Angelman syndrome | GTX-102, knockdown gapmer | Intrathecal | n=74 (enrolling), age 4-17 years, phase 1/2 | Ultragenyx Pharmaceutical | |
NCT05127226 | Angelman syndrome | ION582, knockdown gapmer | Intrathecal | n=51 (enrolling), age 2-50 years, phase 2 | Ionis | |
NCT04849741 | Alexander disease | Zilganersen, knockdown gapmer | Intrathecal, every 12 weeks | n=73 (enrolling), age 2-65 years, phase 1-3 | Ionis | |
NCT05032196 | Huntington’s disease | WVE-003, knockdown gapmer | Intrathecal, every 8 weeks | n=47 (enrolling), age 25-60 years, phase 1b/2a | Wave Life Sciences | |
NCT05686551 | Huntington’s disease | Tominersen knockdown gapmer | Intrathecal, every 16 weeks | n=300 (enrolling), age 25-50 years, phase 2 | Hoffmann-La Roche | |
NCT04165486 | Multiple system atrophy | ION464, knockdown gapmer | Intrathecal | n=40 (enrolling), age 40-70 years, phase 1 | Biogen | |
NCT05575011 | Spinal muscular atrophy | BIIB115/ION306, unknown mechanism | Intrathecal | n=62 (enrolling), age 6 months-55 years, phase 1 | Biogen | |
NCT04768972 | Fused in sarcoma mutations with ALS | Ulfnersen, knockdown gapmer | Intrathecal, every 12 weeks | n=95 (enrolling), age ≥10 years, phase 3 | Ionis | |
NCT04494256 | ALS | BIIB105 (ION541), knockdown gapmer | Intrathecal | n=99 (completed Aug 2024, results unknown), age ≥18 years, phase 2 | Biogen | |
NCT05633459 | ALS | QRL-201, splice modulating | Intrathecal | n=64 (enrolling), age 18-80 years, phase 1 | QurAlis Corporation | |
NCT04539041 | Progressive supranuclear palsy | NIO752, downregulate Tau protein | Intrathecal | n=59 (impending completion), age 40-75 years, phase 1 | Novartis Pharmaceuticals | |
NCT06185764 | Myotonic dystrophy type 1 | VX-670, splice modulating | Intravenous | n=36 (enrolling), age 18-64 years, phase 1/2 | Vertex Pharmaceuticals | |
NCT03913143 | Leber congenital amaurosis 10 | Sepofarsen, splice modulating | Intravitreal, every 4 weeks | n=15 (completed Dec 2023, results unknown), age <8 years, phase 2/3 | ProQR Therapeutics Laboratories Théa | |
NCT04123626 | Retinitis pigmentosa | QR-1123, knockdown gapmer | Intravitreal, every 3 months | n=11 (paused 2023), age ≥18 years, phase 1/2 | ProQR Therapeutics Laboratories Théa | |
NCT05902962 | Retinal dystrophy | VP-001, splice modulating | Intravitreal | n=20 (enrolling), age ≥18 years, phase 1 | PYC Therapeutics |
ASO, antisense oligonucleotide; TANGO, targeted augmentation of nuclear gene output; DEE, developmental and epileptic encephalopathy; ALS, amyotrophic lateral aclerosis.