1. Introduction
Spinal Muscular Atrophy (SMA) is one of the most prevalent autosomal recessive genetic disorders, affecting approximately 1 in 10,000 births[
1]. SMA is mainly characterized by the degeneration of alpha motor neurons located in the anterior horn of the spinal cord and motor nuclei of the lower brainstem resulting in muscle wasting, weakness, hypotonia and difficulties with feeding and respiration[
2,
3,
4,
5].
The underlying cause of classic spinal muscular atrophy (SMA) is typically the homozygous deletion or, less commonly, smaller mutations within the SMN1 gene located on chromosome 5. Additionally, due to intrachromosomal duplication, humans have another paralogous gene SMN2[
6]. However, SMN2 cannot compensate for the loss of SMN1 due to a single-nucleotide polymorphism in exon 7 (C—to—T transition) which creates an intronic splicer silencer N1 (ISS-N1). Splicing modulators can bind to ISS-N1, leading to the exclusion of exon 7 in approximately 90% of SMN2 transcripts. This results in the production of a truncated and unstable protein, SMNΔ7, which is rapidly degraded[
7]. The genetic alterations in SMN1 result in decreased expression of the survival motor neuron protein (SMN). SMN protein is expressed ubiquitously in almost every cell, both in the nucleus and cytoplasm, and it plays a crucial role in various cellular mechanisms. These include the assembly of the spliceosomal machinery, endocytosis, protein translation, and maintenance of cellular homeostasis. Due to its diverse functions and widespread expression, the loss of SMN can result in systemic pathology extending beyond the motor neuron[
8,
9].
Several therapeutic strategies, such as nusinersen, onasemnogene abeparvovec, and risdiplam, have gained regulatory approval by the FDA and EMA for the treatment of SMA. These approaches aim to enhance SMN production either by modifying SMN2 splicing or by replacing the defective SMN1 gene [
10]. Additionally, various other strategies have been explored, including elevating SMN transcript levels, stabilizing SMN protein, implementing neuroprotective measures, employing muscle activators, and more. While these therapies don't directly address the primary deficiency of SMN, they could be used in conjunction with treatments that boost SMN production to offer additional benefits to patients[
11].
This review aims to comprehensively discuss the molecular characteristics, pathophysiology, and currently approved treatments for SMA. Furthermore, we will explore alternative approaches focused on increasing SMN levels.
1.1. Genetic Background of SMA & Diverse Role of SMN
Spinal muscular atrophy (SMA) is caused by deletions or mutations in the survival of motor neuron gene (SMN1), which was originally cloned and characterized by Melki and colleagues[
2]. SMN1, also referred to as SMNT (with T standing for telomere), spans 20 kb and is located in the telomeric region of a 500 kb inverted duplication on chromosome 5q13. This genomic locus is characterized by a high number of repetitions, resulting in instability and frequent deletions in most SMA patients. Unlike other species, such as mice, which have only one copy of the SMN gene, humans have a variable number of centromeric SMN2 genes[
12]. Notably, SMN2 appears to be unique to humans, as chimpanzees have multiple SMN1 copies but no SMN2. SMN2, also referred to as SMNC (with C indicating centromere), shares over 99% nucleotide identity with SMN1. Each SMN gene comprises nine exons, including exons 1, 2a, 2b, 3, 4, 5, 6, 7, and 8 (which encode the 3′ untranslated region (UTR))) responsible for encoding the survival of motor neuron (SMN) protein. However, SMN1 and SMN2 differ by 8 nucleotides, 5 of which are intronic and 3 of which occur in the last 3 exons[
13]. Among these differences, a single functional, coding variant, c.840C>T in exon 7 of SMN2, disrupts an exonic splicing enhancer and simultaneously creates an exonic splicing silencer[
14,
15] (
Figure 1). Normally, SMN1 produces full-length mRNA, which translates into functional SMN protein (294 amino acids, 38 kDa). The SMN1 gene is essential in diverse organisms, in which null mutations are lethal during early development[
16]. In contrast to SMN1, alternative splicing due to the presence of an exonic splicing silencer results in the exclusion of exon 7 in SMN2. This leads to a shortened mRNA that encodes a truncated and unstable SMN protein (SMNΔ7; 282 amino acids, 30.5 kDa), which is rapidly degraded. However, a minority of SMN2 pre-mRNA transcripts retain exon 7 after splicing, leading to the production of approximately 10% of functional protein from SMN2, thereby making it a disease-modifying gene[
17] (
Figure 1).
The SMN protein, translated from the SMN gene, is ubiquitously expressed in all cells and tissues, with particularly high levels in the nervous system, especially the spinal cord[
18]. During gestational and neonatal stages, SMN protein expression is high beyond the neuromuscular system and declines with age, but motor neurons of the spinal cord maintain high SMN levels throughout life[
19,
20]. SMN forms a complex with Gemins2-8 in the cytoplasm and in nuclear bodies called gems, similar to Cajal bodies[
21,
22]. In addition to gemins, other proteins also interact with SMN. These include Sm, Sm-like proteins, RNA helicase A, fibrillarin, GAR1, ribonucleoproteins (RNPs)- heterogenous nuclear RNP U (hnRNP U), hnRNP Q, hnRNP R, and p80-coilin-the marker for Cajal bodies[
23,
24]. The primary role of SMN is in the biogenesis and maintenance of spliceosomal small nuclear ribonucleoprotein (snRNP) assembly, crucial for pre-mRNA splicing. Additionally, SMN is involved in transcriptional regulation, telomerase regeneration, autophagy, cellular trafficking and homeostasis, signal transduction, DNA repair, and recombination[
24].
Structurally, the 294-amino-acid-long SMN protein comprises multiple domains: the N-terminal Gemin2 and nucleic acids binding domain, the central Tudor domain, and the C-terminal proline- and YG box-rich domain. The Tudor domain interacts with coilin protein, a marker of Cajal bodies. The domain also binds to the C-terminal rich arginine- and glycine-rich tails of Sm core proteins to facilitate spliceosome assembly. The YG box is a tyrosine/glycine-rich region in the C-terminus of SMN protein, enabling SMN oligomerization through a glycine zipper structure. Mutations in these domains are linked to SMA, highlighting the importance of SMN's structural integrity for its function[
25].
Reduced expression of SMN protein in SMA leads to impaired α-motor neuron development and degeneration, manifesting as skeletal muscle weakness, the most evident clinical manifestation of SMA[
26]. Early stages of SMA involve impaired motor neuron development, affecting the cell body, axon, and myofibers. This results in delayed synaptic input acquisition, immature firing patterns[
27,
28,
29], reduced motor axon growth and myelination[
30], and deficient NMJ synapse function[
31,
32,
33]. As the disease progresses, terminal motor axons withdraw from NMJ postsynaptic terminals, proximal motor axons degenerate, and motor neuron cell bodies lose synaptic inputs and undergo cell death[
24]. Prolonged denervation of myofibers leads to their replacement by fibro-adipose tissue[
34]. Later stages of SMA involve slower neurodegeneration, primarily affecting distal motor components. Key molecular mediators of neurodegeneration in SMA include the p53 pathway, which is activated due to altered splicing of Mdm2 and Mdm4, as well as JNK signaling, ER stress, and DNA damage[
24].
Ninety-five percent of affected individuals have homozygous deletions of both exons 7 and 8 or only exon 7 of SMN1, regardless of the disease phenotype. Most of the remaining 5% are typically compound heterozygotes with an SMN1 exon 7 deletion and an SMN1 point mutation[
35]. In patients with less severe forms of SMA (types 2 and 3), gene conversion (where SMN1 exon 7 is replaced by SMN2 exon 7) often occurs instead of genuine deletions of SMN1, which are more common in SMA type 1[
36]. The protein domain encoded by exon 7 (amino acids 280–294) is essential for SMN's function[
7,
37]. The SMNΔ7 protein, which lacks this exon, is unstable, rapidly degraded, and deficient in oligomerization[
14,
38] and binding to Sm core proteins[
39], and gem formation[
40]. SMNΔ7 has a significantly shorter half-life compared to full-length SMN due to degradation via the ubiquitin-proteasome system[
41]. However, its stability can be enhanced by coexpressing full-length SMN, which recruits SMNΔ7 into oligomeric complexes[
42]. Additionally, exon 7 includes a cytoplasmic targeting signal vital for transporting SMN into neuronal processes[
43].
Additional intragenic mutations, including missense, nonsense, splice site mutations, insertions, deletions, and duplications, are particularly common in exons 3 and 6 [
44,
45]. These missense mutations are of significant interest as they may alter specific properties of the SMN protein without causing a complete loss of function. Most SMA mutations are clustered in the Tudor domain encoded by exon 3 (e.g., W92S, V94G, G95R, A111G, I116F, Y130C, E134K, and Q136E) and in exon 6, within or near the Y/G box (e.g., L260S, S262G, S262I, M263R, M263T, S266P, Y272C, H273R, T274I, G275S, G279C, and G279V). This mutation distribution indicates that both the Tudor domain and the Y/G box, along with its surrounding regions, are critical for SMN function[
46].
1.2. Clinical Manifestation of SMA
Spinal muscular atrophy (SMA) was first described by Guido Werdnig in 1891 in a case involving muscle weakness in two infant brothers, followed by seven additional cases reported by Johan Hoffmann from 1893 to 1900[
47,
48]. SMA leads to the progressive loss of alpha motor neurons in the ventral spinal cord and motor nuclei of the lower brainstem. Clinically, SMA presents as hypotonia, muscle weakness, and atrophy, with severity varying by genotype. The muscle weakness is predominantly proximal, with greater involvement of the lower extremities, and is usually symmetric, accompanied by diffuse areflexia[
1]. In severe cases, bulbar and respiratory muscle weakness can occur, although facial and ocular muscles are generally spared[
49]. The various phenotypes of spinal muscular atrophy (SMA) were formalized into a classification scheme at a 1991 International Consortium on Spinal Muscular Atrophy, sponsored by the Muscular Dystrophy Association (MDA). This initial classification identified three SMA types based on the highest level of motor function achieved (e.g., sitting or standing) and age of onset. Later modifications further divided the type 3 category by introducing type 4 for adult-onset cases and type 0 for prenatal onset cases resulting in death within weeks[
35,
47] (Table 1).
Table 1.
Classification of Spinal Muscular Atrophy (SMA)[
2,
35,
47,
50,
51,
52,
53].
Table 1.
Classification of Spinal Muscular Atrophy (SMA)[
2,
35,
47,
50,
51,
52,
53].
SMA type |
Age of Onset |
Life Expectancy |
SMN2 Copy Number |
Clinical Manifestation |
Alternative Name |
Estimated SMA Portion |
0 |
Prenatal |
< 1month |
1 |
Require assisted respiration at birth; Fetus dysplay reduced mobement |
- |
Unclear, maybe < 1% |
1 |
0-6 months |
<2 years |
2 |
Unable to sit independently, respiratory and feeding support required |
Werdnig–Hoffman disease |
~60% |
2 |
<18 months |
>2 years |
3, 4 |
Ability to independently sit, inability to walk, respiratory (often non-invasive) and feeding support required |
Dubowitz disease |
~27% |
3a |
18 months – 3 years |
Adult |
3, 4 |
Full ambulation, but slowly progressive muscle atrophy and weakness. |
Kugelberg–Welander disease |
~12% |
3b |
>3 years |
Adult |
4 |
4 |
>21 years |
Adult |
≥4 |
Usually preserved walking ability |
Adult- onset SMA |
~1% |
3. Broader Therapeutic Strategies: SMN-Dependent and Independent Approaches
Research continues to develop drugs for SMA by targeting and increasing SMN expression via splice modulation, leading to the development of additional drugs targeting SMN2.
Branaplam: Branaplam (also known as LMI070), similar to risdiplam, is an orally available, small molecule developed by Novartis as a potential therapy for spinal muscular atrophy (SMA). It was identified through a high-throughput screen designed to find molecules that promote the inclusion of exon 7 in SMN2 pre-mRNA, stabilizing it with splicing factor complexes. In SMA mice, daily administration of Branaplam resulted in a dose-dependent increase in exon 7 inclusion and SMN protein expression, improving body weight and lifespan. The first human phase I/II trial (NCT02268552) for Branaplam began in 2015, targeting SMA patients under six months old with two copies of SMN2. This trial aimed to evaluate the safety, tolerability, and early effectiveness of Branaplam [
11]. However, recruitment was halted in 2016 due to adverse events observed in animal studies while the trial was underway, which included damage to nerves, the spinal cord, testes, and kidney blood vessels. Enrollment resumed in late 2017 with modifications to the study design, including the allowance of both feeding tube and oral administration, and the addition of nerve tests as safety measures. Recruitment was completed in May 2019, encompassing 13 infants in part 1 and 25 infants in part 2. Later that year, the company announced positive progress, with some infants receiving therapy for more than four years, though full results were not disclosed, leaving the complete safety profile unknown (branaplam (LMI070) (smanewstoday.com)). Additionally, clinical deterioration was observed after reducing the subsequent target dose to one-tenth. Due to rapid advancements in SMA treatments, Novartis discontinued Branaplam's development for SMA in mid-2021[
69]. However, Branaplam has shown potential in reducing huntingtin mRNA, the mutated protein in Huntington’s disease, earning FDA Orphan Drug Designation for Huntington's, with a phase IIb trial planned for 2021[
11].
Histone deacetylase inhibitors (HDAC): Histone deacetylase (HDAC) inhibitors have been extensively investigated for their role in treating spinal muscular atrophy (SMA) due to their ability to activate SMN2 transcription by inhibiting deacetylation of chromatin histones, thereby promoting gene expression[
105,
106,
107,
108]. Notably, several HDAC inhibitors, including valproic acid, trichostatin A, and sodium phenylbutyrate (
Figure 2), have demonstrated promising results in increasing SMN levels both in vitro and in vivo[
109]. Valproic acid, a classic class I HDAC inhibitor, has shown beneficial effects in SMA mouse models and patient fibroblasts, leading to its rapid progression to clinical trials[
110]. However, a systematic review of these trials indicated an improvement in motor function but little effect on survival[
111]. Similarly, another class I HDAC inhibitor, phenylbutyrate, was shown to increase SMN protein levels and the number of Gems in SMA fibroblast culture[
108], however a randomized, placebo-controlled trial showed no significant improvement in motor function, leading to the premature termination of its clinical trial (NCT00439569)[
112]. Other small molecules with HDAC inhibitor properties, such as suberoylanilide hydroxamic acid (SAHA/ Vorinostat) [
113], trichostatin A [
114], and resveratrol[
115], also demonstrated success in laboratory models of SMA but have not advanced to clinical trials. While HDAC inhibitors alone do not provide therapeutic benefits equivalent to SMN replacement, they may offer additional neuroprotective support when used in combination with other SMN-targeting therapies[
1,
111]. This concept is supported by recent research demonstrating the advantages of combining the HDAC inhibitor LBH589 (panobinostat) with low doses of Spinraza-like antisense oligonucleotides (ASOs), suggesting a potential synergistic approach for enhancing SMA treatment efficacy[
116].
Neuromuscular Junction targeting therapies: SMA is associated with an impairment of neuromuscular junction dysfunction (NMJ) development, maturation, and function, contributing to muscle weakness and fatigue. Targeting NMJ pathology presents a potential complementary therapy for SMA. Therapeutic strategies targeting NMJ under development include Salbutamol, Amifampridine, NT-1654, and Pyridostigmine [
11,
69] (
Figure 2).
Salbutamol/albuterol, beta- adrenoreceptors agonists, primarily act on Beta2-adrenoreceptors in the smooth muscle of blood vessels, lungs, and intestines. They have shown promise in improving muscle strength by increasing full-length SMN mRNA, SMN protein, and Gem numbers by promoting exon 7 inclusion in SMN2 in vitro and possibly stabilizing acetylcholine receptor clusters at the NMJ[
117,
118]. Although small clinical studies suggest benefits in maintaining motor function or improving respiratory function in SMA type 2 patients, large-scale, placebo-controlled studies are lacking [
69].
Amifampridine, a voltage-dependent K+ channel blocker, enhances neuromuscular transmission and muscle function. Approved for Lambert-Eaton myasthenic syndrome[
11], it was also tested in a randomized, double-blind, placebo-controlled crossover study (NCT03781479) for SMA involving 13 type 3 SMA patients who could walk unaided. The primary outcome was the HFMSE score change, with secondary outcomes including timed tests and quality-of-life assessments. The study indicated that patients receiving amifampridine showed a mean HFMSE improvement of 0.792 compared to the placebo group, with no serious adverse events reported. This suggests that the NMJ could be a potential SMN-independent therapeutic target, highlighting the need for larger studies to confirm amifampridine's role as an adjunctive therapy in SMA treatment[
119].
Pyridostigmine, an acetylcholinesterase inhibitor, slows the degradation of acetylcholine within the synaptic cleft, enhancing cholinergic transmission efficiency. Used to improve muscle strength in myasthenia gravis, an autoimmune disorder that also causes muscle weakness. The SPACE trial (NCT02941328) is a phase 2, monocentre, double-blind, placebo-controlled cross-over trial to assess the efficacy of the pyridostigmine in SMA type II-IV involving 37 participants aged 12 and older. The trial assessed motor fatigue and function using the repeated nine-hole peg test (R9HPT) and the motor function measure (MFM). Results showed no significant difference in R9HPT scores between treatments, and while MFM scores were slightly better with pyridostigmine (average 42.4% vs. 41.6%), the difference was not statistically significant. However, 74.4% of pyridostigmine-treated patients reported less fatigue compared to 29.7% on placebo, indicating a significant reduction in fatigue[
120].
Additionally, subcutaneous administration of NT-1654, the active portion of agrin, has been shown to delay disease progression in SMA mouse models[
121]. The agrin/MuSK signaling pathway, crucial for NMJ formation and maturation, is dysregulated in SMA. Overexpressing agrin or administering agrin-like molecules or downstream mediators like DOK7 can enhance NMJ structure and reduce disease severity in SMA[
122,
123].
Neuroprotective and muscle-enhancing therapies: Recent studies have shown that selectively depleting SMN in the skeletal muscle of mice leads to muscle and neuromuscular junction (NMJ) pathology. It is hypothesized that improving muscle pathology may help preserve proprioceptive synapses on motor neurons, which are typically lost in SMA. Consequently, targeting muscle is seen as a promising therapeutic approach through various strategies (
Figure 2).
Myostatin Inhibitors: Myostatin acts as a negative regulator of muscle growth. Inhibition of the myostatin signaling pathway has shown promising results, especially in less severe models of SMA or in addition to SMN-restoring therapies. Myostatin-deficient animals and humans demonstrate significantly increased musculature[
123,
124].
Apitegromab (SRK-015) is an investigational fully human monoclonal antibody which inhibits the activation of myostatin, thereby preserving muscle mass. In SMA mice, apitegromab improved muscle mass and function[
125]. The Phase 2 TOPAZ clinical trial evaluated the safety and efficacy of apitegromab in nonambulatory patients with SMA types 2 and 3, including 58 participants aged 2 to 21 years. The trial demonstrated sustained improvements in motor function over 24 months, particularly in younger patients given higher doses. The trial's extension phase up to three years confirmed these positive outcomes, with patients showing continued improvement in motor abilities and reduced fatigue. Apitegromab was well-tolerated, with minor adverse effects such as headache, upper respiratory tract infections, pyrexia, nasopharyngitis, cough, and vomiting[
126].
Follistatin is an endogenous antagonist of myostatin; over-expression of recombinant follistatin in SMA mouse muscle leads to increased skeletal muscle mass and survival[
127].
A hybrid activin II receptor (ACTIIR) ligand trap, BIIB110 (ALG 801) inhibits activin receptor type IIB (ActRIIB) ligands, promoting muscle growth, which in turn enhances muscle mass and function. It is currently in phase 1 of clinical development[
104].
Fast Skeletal Muscle Troponin Activators: Reldesemtiv (formally CK-2127107): This drug enhances muscle contractility by prolonging calcium binding to the troponin complex in fast skeletal muscles, reducing the energetic cost of muscle contraction[
128]. In a phase 2 trial (NCT02644668) for SMA type II-IV patients, participants received either 150 mg or 450 mg twice daily for 8 weeks. The higher dose group showed better performance in the 6-minute walk test (6MWT) and the Timed Up and Go (TUG) test, as well as increased maximal expiratory pressure (MEP). Some patients reported serious adverse events such as elevated blood creatine phosphokinase and aspartate aminotransferase levels, along with gastrointestinal infections[
129]. Further studies are required to establish its efficacy, including in type 1 SMA infants.
Neuroprotection: In SMA, neurons and muscles—the primary tissues affected—have high energy demands, making energy pathway targeting potentially neuroprotective and therapeutic[
130]. Olesoxime (TRO19622), a member of the trophos cholesterol-oxime compound family, functions as a mitochondrial pore modulator with neuroprotective properties. Pre-clinical studies indicated its potential to enhance neuron function and survival[
131]. However, a phase 2 placebo-controlled trial in patients with types 2 and 3 SMA revealed only a modest but consistent benefit compared to placebo. A subsequent 18-month follow-up study (OLEOS, NCT02628743) failed to demonstrate significant clinical benefits, leading the pharmaceutical company to halt its development for SMA in June 2018[
132]. Patients from this study were subsequently enrolled in the JEWELFISH (RO7034067) clinical trial with risdiplam[
69].
Other potentially neuroprotective agents, such as riluzole and gabapentin, have been explored for their effects on treating SMA by targeting excitotoxicity. Riluzole, known for its modest effects in ALS, was tested in a small preliminary phase I trial involving seven participants, which suggested some potential benefit[
133]. Subsequently, a phase II/III multicenter, randomized, double-blind study was conducted to assess riluzole's efficacy and safety in young adults with types 2 and 3 SMA (NCT00774423). However, the majority of results were not encouraging, and the studies failed to demonstrate significant efficacy[
109,
134]. Similarly, gabapentin was tested in type 2/3 SMA patients due to its effects in ALS. While one study showed some improvement in motor function, another did not demonstrate any significant benefit[
135,
136].
Targeting cell death mechanism: In SMA, neurodegeneration is linked to disruptions in core pathways of cell homeostasis, including autophagy, ubiquitin homeostasis, and apoptotic pathways[
137,
138]. Therapeutic strategies targeting molecules involved in these pathways are under investigation. Celecoxib (
Figure 2), a selective cyclooxygenase-2 (COX-2) inhibitor that can cross the blood-brain barrier, has shown potential in increasing SMN protein levels through the activation of the p38 pathway in SMA cell and rodent models[
139]. However, despite these promising preclinical findings, a phase II clinical trial of celecoxib for SMA was prematurely terminated, and the results have not yet been published (NCT02876094)[
11].
Targeting cytoskeleton: In SMA, the upregulation of the RhoA/Rho kinase (ROCK) pathway disrupts actin dynamics, which disrupts cytoskeleton structure and hinders neuronal growth and regeneration[
140]. Therefore, targeting this pathway can be a possible therapeutic option. Y-27632[
141] and the FDA-approved fasudil[
142], pharmacological inhibitors of ROCK pathway (
Figure 2), have demonstrated improvements in survival, neuromuscular junction (NMJ) maturation, and muscle development in SMA mouse models.
4. Challenges in the treatment era: Are We There Yet in the Battle Against SMA?
The therapeutic landscape for Spinal Muscular Atrophy (SMA) has seen remarkable advancements with significant progress in extending patient lives and improving motor function. Despite the success of FDA-approved treatments like nusinersen, onasemnogene abeparvovec, and risdiplam in improving patient outcomes, these therapies come with several challenges.
Nusinersen, for instance, must be injected directly into the cerebrospinal fluid (CSF) intrathecally to ensure it reaches the CNS to treat lower motor neurons, although this method is often associated with thrombocytopenia and injection-site adverse events[
55,
143,
144]. Adult SMA patients face additional challenges such as scoliosis, spine or thorax deformities, and prior spinal surgeries, which complicate the safe administration of nusinersen[
145]. Furthermore, intrathecal administration limits nusinersen's efficacy to the central nervous system, failing to address SMA's multi-organ impact effectively[
146]. Recent studies in animal models and humans indicate that SMA affects multiple organs, including the heart, peripheral nervous system, skeletal muscle, liver, and vasculature[
5,
147]. For example, muscle-specific SMN loss in SMA mouse models results in compromised motor performance, premature death, muscle fiber defects, and neuromuscular junction abnormalities. In human fetuses with SMA, SMN deficiency causes delayed growth and maturation of myotubules[
148]. Untreated severe SMA patients with only one copy of SMN2 have shown thrombotic occlusions of small blood vessels, leading to digital necrosis, suggesting that severe SMN deficiency may also manifest as a vascular disease[
149]. The increasing evidence of multi-organ involvement in SMA suggests that ASO therapies targeting motor neurons alone may be insufficient for long-term management of SMA pathology. Additionally, a recent study[
150] investigating nusinersen's biodistribution in post-mortem infant spinal cord samples found variability in ASO distribution, with notably lower concentrations in the cranial portion of the spinal cord and in the brain suggesting uneven distribution within the CNS and potentially affecting treatment efficacy. Despite positive outcomes from pre-symptomatic intrathecal treatment in the NURTURE trial, the long-term effects and efficacy of nusinersen remain uncertain[
66,
67]. Furthermore, gaps remain in understanding whether nusinersen helps achieve significant milestones, regain function, or avoid serious side effects and ventilation support. More research and long-term data are needed to justify its use in specific SMA patient subgroups.
Onasemnogene abeparvovec (Zolgensma) is a one-time gene therapy that offers significant survival benefits for SMA patients. However, it raises concerns about liver toxicity and reduced efficacy over time[
151,
152]. This reduction happens because cell division dilutes the episomal SMN cDNA within the AAV vector, limiting expression to non-dividing cells like neurons[
11]. The body's immune response to the viral vector complicates repeated treatments, while systemic delivery and maintenance of the treatment is crucial for long-term benefits due to the involvement of peripheral organs in the disease. Onasemnogene has to be administered within the first two years of life, making newborn screening for SMA crucial for early diagnosis and timely treatment, given the limited window for effective intervention. Serious side effects, such as liver damage, thrombotic microangiopathy, thrombocytopenia, and cardiac toxicity has also been observed for onasemnogene and therefore require careful patient monitoring. Furthermore, recent studies have highlighted a toxic gain of function of SMN, especially in the sensorimotor circuit, including the loss of proprioceptive neurons. Similar results were also found when intrathecal injection was explored as an alternative delivery method (NCT03381729). Pre-clinical trials involving intrathecal injection have shown concerns about neurotoxicity, including ataxia, loss of proprioceptive synapses, neuroinflammation, and neurodegeneration[
153,
154,
155]. These issues highlight the need for more long-term data to fully understand the therapy's effectiveness and safety.
Risdiplam, an oral medication, provides a more accessible treatment option, especially for older patients after receiving its approval in 2020 for all SMA types in patients 2 years of age. Although it overcomes the necessity for invasive intrathecal injections, its long-term effects are still unknown. Potential risks include male fertility impairment, growth issues, retinal toxicity, and off-target effect although these have not yet been observed in humans[
102,
156,
157]. The drug is contraindicated for patients with hepatic abnormalities, which could limit its use in many older SMA patients who often present with liver issues.
Apart from these, the extremely high cost of these treatments poses a significant barrier to widespread accessibility. While early treatment is critical for optimal outcomes[
158], there remains a subset of patients who do not respond to these therapies[
66]. The variability in response can be attributed to factors beyond SMN1 and SMN2 genes, including other genetic factors, environmental influences, and disparities in medical care quality and accessibility.
5. Future directions: Advancing the battle against SMA
The field of Spinal Muscular Atrophy treatment has significantly evolved with the introduction of nusinersen, onasemnogene abeparvovec, and risdiplam, which have become the standard of care. Despite their ground-breaking nature, it has become clear through long-term follow-ups that SMN2 gene modulation or SMN1 gene replacement alone does not constitute a cure. Future directions in SMA treatment involve incorporating SMN-independent therapies alongside SMN-dependent ones to provide broader benefits, especially for patients diagnosed later or with milder forms of SMA. A cross-disease approach, repurposing drugs with established safety profiles, could also expedite the development of effective therapies. Additionally, combining currently approved therapies has garnered interest due to their different approaches to treating SMA. For example, Lee et al. provided insights on patients first treated with nusinersen followed by onasemnogene[
159], while another small study involving five patients treated with both therapies showed phenotype improvements but highlighted potential liver toxicity risks[
160]. More research is needed to determine the therapeutic benefits and cost-benefit analysis of combination treatments.
In addition to drug-based therapies, a multidisciplinary approach is essential for effective SMA treatment, incorporating physiotherapy, rehabilitation, respiratory management, orthopedic care, and nutritional support[
161]. Further research is needed to determine the most effective combination of these treatments.
Recent evidence strongly supports that early intervention provides greater clinical benefits than treating symptomatic patients, with a high potential to achieve age-appropriate motor milestones[
158]. Therefore, neonatal genetic screening is crucial for early diagnosis and treatment. In Belgium, scientists completed an observational newborn screening study titled "Sun May Arise on SMA"[
162]. Similarly, Prof. Servais launched a newborn screening study in Oxford, UK, in March 2022, expected to conclude in August 2025[
163].
Finally, researching to improve existing treatment approaches for SMA is essential to offer more effective, safer, and accessible options. One promising area of research focuses on enhancing the efficacy and minimizing the toxicity of antisense oligonucleotides (ASOs) as nusinersen's success highlights the promise and efficiency of ASO-based therapies, yet there is room for enhancement. A notable advancement is the development of phosphorodiamidate morpholino oligomers (PMOs), a different chemistry of ASO, which are neutrally charged due to their phosphorodiamidate linkage, offer enhanced stability and minimal toxicity[
164]. These properties have made PMOs effective therapeutic options, as evidenced by the approval of four PMO-based ASOs for Duchenne Muscular Dystrophy (DMD)[
165]. In the context of SMA, studies on mouse models have demonstrated that PMOs can significantly increase SMN2 levels and improve survival rates through various delivery routes, including intrathecal and intravenous injections. Although the superiority of these delivery routes remains debated. Most notably, PMOs have shown lower toxicity, prolonged survival, and most importantly, effective SMN restoration in central nervous system (CNS) tissues. Additionally, PMOs' neutral charge allows them to be conjugated with cell-penetrating peptides (CPPs) for improved delivery and uptake in target tissues[
166,
167,
168]. CPPs, short sequences that facilitate the uptake of cargo into target cells, were discovered in the late 1980s[
165]. In the context of SMA, CPPs such as Pip6a, RXR peptides, and DG9 have shown promising results by enhancing cellular uptake and crossing the blood-brain barrier, leading to better restoration of SMN levels, prolonged survival, and improved phenotypes in mouse models[
169,
170,
171]. Overall, it is crucial to continue researching ways to enhance current treatment approaches while exploring new options to ensure the best possible outcomes for SMA patients.