1258984-36-9
Spinal Muscular Atrophy (SMA)
Nusinersen, marketed under the brand name Spinraza®, is a significant therapeutic agent developed for the treatment of spinal muscular atrophy (SMA).[1] SMA is a rare, autosomal recessive neuromuscular disorder characterized by the progressive degeneration of alpha motor neurons in the spinal cord, leading to muscle weakness, atrophy, and, in its most severe forms, premature death.[3] The underlying cause of SMA is a deficiency of the Survival Motor Neuron (SMN) protein due to mutations or deletions in the SMN1 gene.[3]
The approval of Nusinersen by the U.S. Food and Drug Administration (FDA) in December 2016 marked a pivotal moment in the management of SMA, as it was the first therapy specifically targeting the molecular basis of the disease to receive such approval.[2] This development represented a paradigm shift, moving beyond purely supportive care to a disease-modifying intervention capable of altering the natural history of SMA. For many patients and families affected by this devastating condition, Nusinersen brought forth new hope by offering the potential to improve motor function, enhance survival, and positively impact quality of life.[2] Prior to its availability, the prognosis for individuals with severe SMA was grim, with management focused on palliative measures and respiratory support.[2]
Nusinersen is classified as a biotech drug and is an antisense oligonucleotide (ASO).[2] Its DrugBank identification number is DB13161, and its Chemical Abstracts Service (CAS) number is 1258984-36-9.[1] The development and approval of Nusinersen as an orphan drug in multiple jurisdictions underscore the specific regulatory frameworks and incentives established to foster the creation of treatments for rare diseases.[4] SMA's low prevalence qualifies it for such designations, which often include benefits like market exclusivity and research support.[4] These incentives are critical for encouraging investment in therapies for smaller patient populations, where the conventional pharmaceutical development model faces challenges in recouping substantial research and development expenditures. This context is fundamental to understanding both the journey of Nusinersen to market and the subsequent discussions surrounding its cost and accessibility.
Nusinersen is a synthetic antisense oligonucleotide meticulously designed for therapeutic use. Its chemical structure features key modifications to enhance its stability, binding affinity, and pharmacokinetic profile. Specifically, the 2'-hydroxy groups of the ribofuranosyl rings within the oligonucleotide chain are replaced with 2'-O-(2-methoxyethyl) (2'-MOE) groups, and the phosphate linkages that form the backbone of the molecule are substituted with phosphorothioate linkages.[8] These chemical alterations are not merely incidental; they are integral to Nusinersen's efficacy as an ASO. Unmodified oligonucleotides are typically susceptible to rapid degradation by endogenous nucleases and exhibit poor cellular uptake. The 2'-MOE modification confers increased resistance to nuclease activity and enhances the ASO's binding affinity to its target RNA sequence. Similarly, the phosphorothioate backbone further improves nuclease resistance and can favorably alter the drug's distribution and lifespan within the body.[8]
The molecular formula of Nusinersen is C234H340N61O128P17S17, and it has a molecular weight of approximately 7127.19 g/mol.[3] Its exact mass is 7124.282 Da.[3] In its solid form, Nusinersen appears as a white to off-white powder.[3] The specific sequence of Nusinersen is RNA, 2'-O-(2-methoxyethyl)(m5U-m5C-A-m5C-m5U-m5U-m5U-m5C-A-m5U-A-A-m5U-G-m5C-m5U-G-G).[15]
For clinical administration, Nusinersen (Spinraza®) is supplied as a sterile, preservative-free, clear, and colorless solution intended for intrathecal injection. Each vial contains 12 mg of Nusinersen in a 5 mL volume, corresponding to a concentration of 2.4 mg/mL.[2] The pH of the solution is approximately 7.2.[16] The choice of formulation as a solution for direct intrathecal injection is a direct consequence of Nusinersen's pharmacokinetic properties. As an ASO, it is a relatively large and charged molecule, which typically limits its ability to cross the blood-brain barrier effectively if administered systemically.[17] Intrathecal administration bypasses this barrier, allowing the drug to reach its target tissues within the central nervous system (CNS).
Spinal muscular atrophy arises from the loss of functional Survival Motor Neuron (SMN) protein, predominantly due to homozygous deletion or mutation of the SMN1 gene.[3] While humans possess a nearly identical gene, SMN2, a critical single nucleotide difference (a C-to-T transition within exon 7) distinguishes it from SMN1. This alteration disrupts a key splicing enhancer site, leading the SMN2 gene to primarily produce an alternatively spliced mRNA transcript that excludes exon 7. The resultant protein, known as SMN$\Delta$7, is unstable and rapidly degraded, and is not fully functional.[3] Consequently, only a minor fraction (approximately 10-15%) of the protein produced from the SMN2 gene is full-length, functional SMN protein.[7] The amount of functional SMN protein produced from SMN2 is insufficient to fully compensate for the absence of SMN1-derived protein, leading to the motor neuron degeneration characteristic of SMA.
Nusinersen is an 18-mer antisense oligonucleotide specifically engineered to address this SMN protein deficiency.[8] It functions as a splice-altering oligonucleotide. The core of Nusinersen's mechanism lies in its ability to modulate the splicing of SMN2 pre-messenger RNA (pre-mRNA). It is designed to bind with high specificity to a sequence within intron 7 of the SMN2 pre-mRNA, known as Intronic Splicing Silencer N1 (ISS-N1).[6] The binding of Nusinersen to ISS-N1 is thought to displace or prevent the binding of certain splicing inhibitory proteins, such as heterogeneous nuclear ribonucleoproteins (hnRNPs), thereby antagonizing the silencing effect of this site.[8] This steric blockade effectively unmasks or enhances the recognition of the proper splicing signals for exon 7.
By altering the balance of splicing factors at this critical juncture, Nusinersen promotes the inclusion of exon 7 into the mature SMN2 mRNA transcript.[2] This "corrected" SMN2 mRNA is then translated into a greater quantity of full-length, functional SMN protein.[4] The therapeutic strategy of Nusinersen, therefore, does not involve replacing the missing SMN1 gene, as in gene therapy approaches. Instead, it ingeniously leverages the existing SMN2 gene, effectively converting this "backup" gene into a more robust producer of the essential SMN protein. This increased production of SMN protein within motor neurons is believed to support their survival and function, thereby mitigating the neuromuscular degeneration and alleviating the symptoms associated with SMA.[4] The precision of this ASO, designed to target a specific sequence within SMN2 pre-mRNA, exemplifies the targeted nature of oligonucleotide therapeutics, aiming to maximize efficacy while minimizing potential off-target effects.
The pharmacodynamic effects of Nusinersen are consistent with its mechanism of action, focusing on the increased production of functional SMN protein and subsequent clinical benefits. In vitro assays and studies conducted in transgenic animal models of SMA have demonstrated that Nusinersen effectively increases the inclusion of exon 7 in SMN2 mRNA transcripts, leading to a corresponding rise in the production of full-length SMN protein.[8] The primary physiological effect of Nusinersen is achieved through this increased protein synthesis.[12]
Crucially, evidence of this mechanism has also been observed in humans. Autopsy samples obtained from a small number of SMA patients (n=3) treated with Nusinersen revealed higher levels of SMN2 mRNA containing exon 7 in the thoracic spinal cord compared to samples from untreated SMA infants.[8] Although the sample size is limited, these findings provide vital human evidence that Nusinersen reaches its intended target tissue within the CNS and exerts its anticipated molecular effect, validating the therapeutic hypothesis in patients.
Regarding cardiac electrophysiology, concerns about potential effects on QT interval prolongation, a common area of investigation for new drugs, have been addressed. In a study involving 121 patients with SMA who received either Nusinersen or a sham control, QTcF values greater than 500 ms and changes from baseline values greater than 60 ms were observed in 5% of patients receiving Nusinersen. However, it is important to note that, compared to the sham-control group, there was no associated increase in the incidence of cardiac adverse reactions linked to delayed ventricular repolarization in patients treated with Nusinersen.[8] This suggests that while minor electrocardiographic changes may occur in a small subset of patients, they do not appear to translate into clinically significant cardiac adverse events. Nevertheless, this observation warrants ongoing vigilance in clinical practice.
The pharmacokinetic profile of Nusinersen is characterized by its unique route of administration and its behavior within the cerebrospinal fluid (CSF) and systemic circulation.
Absorption and Distribution:
Nusinersen is administered via intrathecal injection directly into the CSF by lumbar puncture.2 This route is necessary because, like many ASOs, Nusinersen does not efficiently cross the blood-brain barrier if administered systemically.17 Following intrathecal administration, Nusinersen distributes from the CSF to the target tissues within the CNS, including the spinal cord and brain.8 It also distributes to peripheral tissues to some extent.13
Trough plasma concentrations of Nusinersen are relatively low compared to the concentrations achieved in the CSF, reflecting its primary distribution within the CNS after intrathecal delivery. The median time to maximum plasma concentration (Tmax) has been reported to range from 1.7 to 6.0 hours. Mean plasma Cmax (maximum concentration) and AUC (area under the curve) values increase approximately dose-proportionally up to the clinical dose of 12 mg.[8] The estimated volume of distribution is approximately 0.4 L in the CSF and significantly larger in plasma, at approximately 29 L.[8] This pharmacokinetic profile, particularly the low plasma versus high CSF concentrations and the specific volume of distribution for the CSF, underscores the rationale for intrathecal administration to achieve therapeutic drug levels in the CNS. High plasma protein binding (>94%) would further limit CNS availability if given systemically.
Protein Binding:
Nusinersen exhibits differential protein binding in CSF and plasma. In the CSF, protein binding is low, at less than 25%. In contrast, plasma protein binding is high, exceeding 94%.8
Metabolism:
Nusinersen is primarily metabolized through hydrolysis mediated by exonucleases (both 3'- and 5'-exonucleases).5 This enzymatic degradation leads to the formation of chain-shortened oligonucleotides, which are generally considered pharmacologically inactive.8 Metabolites such as N-1 (shortened by one nucleotide) can be detected in the CSF, while more extensively shortened metabolites (e.g., N-1, N-2, N-3) are predominantly found in plasma.8 Some depurination and depyrimidination products have also been identified.20 Significantly, Nusinersen is not a substrate for, nor an inhibitor or inducer of, cytochrome P450 (CYP450) enzymes.5 This characteristic is advantageous as it implies a low likelihood of pharmacokinetic drug-drug interactions mediated by the CYP450 system, a common pathway for many other medications. This simplifies co-administration with other drugs, which is relevant for SMA patients who may have comorbidities.
Route of Elimination:
The primary route of elimination for Nusinersen and its metabolites is via urinary excretion.5
Half-life and Clearance:
Nusinersen exhibits a long terminal elimination half-life. In the CSF, the mean terminal elimination half-life is estimated to be between 135 and 177 days. In plasma, it is estimated to be between 63 and 87 days.5 The drug demonstrates slow clearance from the body.8 This extended half-life in the CSF is a key pharmacokinetic feature that supports the maintenance dosing interval of every four months, as the drug persists in the target compartment for a considerable period. The initial loading doses are designed to rapidly achieve steady-state concentrations.
It has been noted that body weight is the only patient variable known to significantly affect the pharmacokinetics of Nusinersen.[17]
Nusinersen (Spinraza®) has received regulatory approval in numerous countries worldwide, reflecting a global recognition of its therapeutic value in treating spinal muscular atrophy.
In the United States, Nusinersen was approved by the Food and Drug Administration (FDA) on December 23, 2016, under the brand name Spinraza®.[2] This landmark approval made it the first drug authorized for the treatment of SMA in both pediatric and adult patients.[2] The FDA indication covers all ages and types of SMA.[2] The development and review process was expedited, with Biogen completing its rolling New Drug Application (NDA) submission on September 26, 2016, and the FDA (along with the European Medicines Agency) accepting the applications for review on October 28, 2016.[9] The FDA granted Nusinersen Fast Track designation, Priority Review status, and Orphan Drug designation.[13] These accelerated pathways are typically reserved for therapies that address serious conditions and demonstrate the potential to fill an unmet medical need or offer significant improvements over existing treatments. The relatively swift progression from NDA submission to approval highlights the urgency and the promising nature of the clinical data supporting Nusinersen. The drug is marketed by Biogen Inc. and was co-developed by Ionis Pharmaceuticals and Biogen.[2]
The European Medicines Agency (EMA) granted marketing authorization for Spinraza® on May 30, 2017.[12] The approved indication in the European Union is for the treatment of 5q spinal muscular atrophy, which refers to SMA caused by a genetic defect on chromosome 5q.[4] Nusinersen had previously received an orphan medicine designation from the EMA on April 2, 2012, recognizing the rarity of SMA.[4] The marketing authorization holder in the EU is Biogen Netherlands B.V..[12] The EMA, in its assessment, acknowledged the serious nature of SMA and the critical need for effective treatments, concluding that Spinraza® provides clinically meaningful improvements for patients.[4] The EMA's approval shortly after the FDA's, along with a similar orphan drug status, signaled a strong international consensus on the favorable benefit-risk profile of Nusinersen for SMA.
Health Canada approved Spinraza® (nusinersen) on June 29, 2017, for the treatment of 5q Spinal Muscular Atrophy.[11] The manufacturer for the Canadian market is Biogen Canada Inc..[11] Initially, the New Drug Submission (NDS) was filed under Health Canada's Notice of Compliance with Conditions (NOC/c) policy, which allows earlier market access for promising drugs treating serious conditions. However, upon completion of the review, the available clinical data were deemed sufficient to support a full Notice of Compliance (NOC).[11] The Health Canada approval in June 2017 covered all pre-symptomatic, type I, II, and III SMA patients, irrespective of age.[10] Subsequently, efforts were made to secure public funding. For instance, in Ontario, publicly funded access was expanded in June 2019 to include pre-symptomatic patients and those with Type I, II, and III SMA under the age of 18, following an earlier decision in November 2018 for Type I SMA.[10] This progression illustrates the multi-stage process often required to make innovative, high-cost medications broadly accessible within public healthcare systems.
The Therapeutic Goods Administration (TGA) of Australia approved Spinraza® on November 3, 2017, for the treatment of all individuals with 5q SMA.[7] Following TGA approval, reimbursement on the Pharmaceutical Benefits Scheme (PBS) has been expanded incrementally. In 2018, PBS listing included pediatric patients (18 years and younger) with infant or childhood onset of SMA who demonstrated symptoms before the age of three.[7] Spinraza® was also funded for the presymptomatic treatment of babies diagnosed with SMA who have one or two copies of the SMN2 gene, and for adults diagnosed with SMA whose symptoms appeared before 19 years of age.[7] Most recently, effective March 1, 2024, the PBS listing was further expanded to include reimbursement for presymptomatic babies with three copies of the SMN2 gene.[7] This phased expansion of PBS reimbursement likely reflects the accumulation of clinical evidence, such as data from the NURTURE trial (which demonstrated benefits in presymptomatic infants with varying SMN2 copy numbers), influencing policy decisions to broaden access, particularly for early intervention strategies.[22]
In Japan, Spinraza® was approved by the Pharmaceuticals and Medical Devices Agency (PMDA) on July 3, 2017.[13] The indication is for the treatment of infantile spinal muscular atrophy or, more broadly, SMA associated with a mutation in the SMN1 gene.[13] The marketing authorization holder in Japan is Biogen Japan.[13] A re-examination period of 10 years was set, which is a standard PMDA practice for new drugs to ensure long-term monitoring of safety and efficacy.[25] Approval in Japan, a major pharmaceutical market with rigorous regulatory standards, further cemented Nusinersen's global acceptance as a key therapy for SMA.
Table 1: Summary of Key Regulatory Approvals for Nusinersen (Spinraza®)
Regulatory Agency | Approval Date | Approved Indication(s) | Key Notes |
---|---|---|---|
U.S. FDA | Dec 23, 2016 | Spinal Muscular Atrophy (SMA) in pediatric and adult patients (all ages and types) | Orphan Drug, Fast Track, Priority Review 2 |
EMA (European Union) | May 30, 2017 | 5q Spinal Muscular Atrophy | Orphan Medicine (designated Apr 2, 2012) 4 |
Health Canada | June 29, 2017 | 5q Spinal Muscular Atrophy (all pre-symptomatic, type I, II, III SMA patients) | Full Notice of Compliance (NOC) 10 |
TGA (Australia) | Nov 3, 2017 | 5q Spinal Muscular Atrophy (all people) | PBS reimbursement expanded over time for various patient subsets 7 |
PMDA (Japan) | July 3, 2017 | Infantile Spinal Muscular Atrophy / SMA associated with SMN1 gene mutation | 10-year re-examination period 13 |
The clinical development program for Nusinersen has provided robust evidence of its efficacy across the spectrum of SMA types and patient ages, fundamentally altering the natural history of the disease.
The ENDEAR trial was a pivotal Phase 3, multicenter, randomized, double-blind, sham-procedure controlled study that evaluated Nusinersen in 122 infants with symptomatic infantile-onset SMA (consistent with SMA Type 1) who were 7 months of age or younger at screening.[2] The primary endpoints focused on motor milestone response, assessed using the Hammersmith Infant Neurological Examination (HINE), and event-free survival, defined as the time to death or the use of permanent assisted ventilation (≥16 hours/day continuously for >21 days in the absence of an acute reversible event or tracheostomy).[2]
The results of the ENDEAR trial were transformative. An interim analysis revealed a statistically significant and clinically meaningful benefit in the Nusinersen-treated group, with 41% of infants achieving a motor milestone response compared to 0% in the sham-control group.[2] This profound difference led to the early termination of the trial, allowing patients in the sham group to transition to active treatment.[26] The final analysis, including 121 patients, confirmed these findings: 51% of infants in the Nusinersen group achieved a motor milestone response (e.g., head control (22%), ability to roll over (10%), ability to sit independently (8%), and ability to stand (1%)) versus 0% in the sham-control group.[2]
Furthermore, Nusinersen treatment resulted in a 47% reduction in the risk of death or permanent ventilation compared to the sham-control group (hazard ratio 0.53; 95% CI, 0.32-0.89; p=0.005).[2] A separate analysis showed a 63% lower risk of death alone in the Nusinersen group (HR 0.37; 95% CI, 0.18-0.77; p=0.004).[27] Significant improvements were also observed in the Children’s Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP INTEND) scores, with 71% of Nusinersen-treated infants achieving a response (defined as an increase of ≥4 points from baseline) compared to 3% in the sham group.[26] These results were groundbreaking, demonstrating for the first time that a pharmacological intervention could substantially alter the devastating trajectory of infantile-onset SMA, a condition where historically most infants did not survive beyond two years of age or achieve any major motor milestones.
Subgroup analyses from the ENDEAR study suggested that infants with a shorter disease duration at the time of treatment initiation (≤13.1 weeks) experienced a greater likelihood of event-free survival.[27] This finding underscores the critical importance of early diagnosis and intervention to maximize therapeutic benefits, as treatment initiated before extensive motor neuron loss is more likely to preserve function and facilitate developmental progress.
The long-term efficacy and safety of Nusinersen in patients from the ENDEAR trial (and other feeder studies) are being evaluated in the SHINE open-label extension study (NCT02594124). Data from SHINE suggest that the motor function improvements or stabilization observed with Nusinersen can be maintained for up to nearly 6 years of treatment.[28] This durability of effect is vital for a chronic condition like SMA, providing reassurance about the long-term value of continuous Nusinersen therapy.
The CHERISH trial was a Phase 3, multicenter, randomized, double-blind, sham-procedure controlled study that assessed the efficacy and safety of Nusinersen in 126 children with later-onset SMA (symptom onset after 6 months of age; ages 2 to 12 years at enrollment; non-ambulatory, consistent with SMA Type 2 or non-ambulatory Type 3).[2] The primary endpoint was the change from baseline in the Hammersmith Functional Motor Scale Expanded (HFMSE) total score at 15 months of treatment.[29]
The CHERISH trial also demonstrated statistically significant and clinically meaningful benefits for Nusinersen. At 15 months, children treated with Nusinersen showed a mean improvement of 3.9 points from baseline on the HFMSE, whereas children in the sham-control group experienced a mean decline of 1.0 point. This resulted in a highly significant mean treatment difference of 4.9 points in favor of Nusinersen.[2] Overall, 57% of children receiving Nusinersen showed improvement in HFMSE scores compared with 26% of children receiving placebo.[4] These results indicate Nusinersen's ability not only to stabilize but also to improve motor function in children with later-onset SMA, who typically face a progressive decline in motor abilities. Data from other endpoints, including the attainment of new motor milestones and improvements in upper limb motor function as measured by the Revised Upper Limb Module (RULM), were consistently in favor of the children who received Nusinersen.[29]
Patients from the CHERISH study were also eligible to enroll in the SHINE extension study. Long-term follow-up data from SHINE, encompassing patients from CHERISH and other studies (CS2/CS12), suggest sustained improvements or stabilization in HFMSE, ULM, and the 6-Minute Walk Test (6MWT) for up to nearly 6 years.[28] For instance, in the CS2/CS12 studies, patients with later-onset SMA showed mean HFMSE score improvements of 10.8 points (for those consistent with Type 2) and 1.8 points (for those consistent with Type 3) by day 1150.[28]
Beyond objective motor function scales, the CHERISH trial also explored broader impacts. Analyses indicated that treatment with Nusinersen was associated with a reduction in caregiver burden (measured by the Assessment of Caregiver Experience with Neuromuscular Disease - ACEND) and improvements in health-related quality of life (HRQoL) for both patients and parents (measured by the Pediatric Quality of Life Inventory - PedsQL) compared to sham control.[30] Improvements in domains such as feeding/grooming/dressing, transfer, and mobility translate to greater independence and reduced reliance on caregivers, representing meaningful outcomes for families affected by SMA.
While group-level data from CHERISH were positive, it was noted that the response to Nusinersen treatment varied among individuals. Factors such as the age at symptom onset and the time to the first dose were identified as influencing factors, but a considerable degree of inter-patient variability in treatment response was not fully explained by these elements.[17] This suggests that other individual patient characteristics, such as SMN2 copy number, baseline disease severity, or the presence of genetic modifiers, likely contribute to the observed differences in outcomes.
The NURTURE trial (also known as CS5) is an ongoing Phase 2, open-label study evaluating Nusinersen in presymptomatic newborns who are genetically diagnosed with SMA and are highly likely to develop SMA Type 1 or Type 2 (based on having two or three copies of the SMN2 gene). A critical aspect of this study is early intervention, with all 25 participants receiving their first dose of Nusinersen before 6 weeks of age.[2] The primary aim is to assess the efficacy and safety of Nusinersen in preventing or delaying the onset of SMA symptoms and the need for respiratory intervention or death.[6]
The results from the NURTURE trial have been particularly compelling, demonstrating the profound impact of presymptomatic treatment. As of the latest published data (analyzed after approximately 5 years of treatment, July 2023), all 25 participants were alive and free from the need for permanent ventilation.22 Participants continued to maintain and achieve World Health Organization (WHO) motor milestones, with many reaching them within normal developmental timeframes.22
Key motor milestone achievements include:
Children with three copies of the SMN2 gene generally achieved all expected WHO motor milestones, with nearly all milestones met within normal developmental windows.[22] CHOP INTEND scores also showed significant improvement, with 22 out of 25 participants achieving the maximum score on the scale.[22] Average scores on the HFMSE have continued to improve over the duration of the study.[22]
The NURTURE data strongly suggest that initiating Nusinersen treatment before the onset of clinical symptoms can lead to motor development outcomes that are unprecedented for SMA, approaching normal development, particularly for children with three SMN2 copies. These outcomes stand in stark contrast to the natural history of untreated SMA Type 1 (which many infants with two SMN2 copies would develop), where milestones such as sitting or walking are typically never achieved, and survival is severely limited. The trial also highlights the influence of SMN2 copy number on outcomes even with treatment; while all presymptomatically treated infants benefit substantially, those with three SMN2 copies tend to achieve milestones more consistently and within typical developmental periods compared to those with two copies. The remarkable success of presymptomatic treatment in the NURTURE study provides the most robust scientific rationale for universal newborn screening for SMA, as this allows for diagnosis and initiation of therapy during the optimal window before irreversible motor neuron loss occurs. The NURTURE trial has been extended to evaluate the longer-term efficacy and safety of Spinraza® for up to 8 years of age.[22] Throughout the study, Nusinersen has been well-tolerated, with no new safety concerns identified during the extended follow-up.[22]
While the pivotal registration trials for Nusinersen primarily focused on pediatric populations, the FDA approval includes adult patients.[2] The evidence base for Nusinersen's efficacy in adults has been growing through real-world observational studies.
A prospective, non-interventional, multicenter observational cohort study conducted in Germany included adult patients with 5q SMA (ages 16-65 years).[33] Data analysis from 124 patients at 6 months, 92 at 10 months, and 57 at 14 months of Nusinersen treatment showed statistically significant mean increases in HFMSE scores compared to baseline at all time points (e.g., a mean difference of +1.73 points at 6 months and +3.12 points at 14 months). Clinically meaningful improvements, defined as an increase of ≥3 points on the HFMSE, were observed in 28% of patients at 6 months, 35% at 10 months, and 40% at 14 months.[33]
More recently, a larger prospective European multinational observational study involving 237 adult SMA patients from Germany, Switzerland, and Austria provided longer-term data up to 38 months.[34] This study reported significant mean increases from baseline in HFMSE scores (mean difference +1.52 points at 38 months), RULM scores (mean difference +0.72 points at 38 months), and 6MWT distance (mean difference +32.20 meters at 38 months). These improvements or stabilizations were sustained over the 38-month follow-up period.[34]
These real-world observational studies are crucial as they provide evidence supporting the use of Nusinersen in adult SMA patients, a population not included in the initial pivotal trials. The natural history of SMA in adults is often characterized by a slow, progressive decline in motor function or a period of stabilization followed by decline. Therefore, the observed improvements or sustained stabilization in motor function scores (HFMSE, RULM, 6MWT) in a substantial proportion of treated adults represent a positive deviation from this expected trajectory. For adult patients, who may have lived with functional limitations for many years, even modest objective improvements or the halting of disease progression can translate into meaningful benefits in terms of daily activities, independence, and overall quality of life. The definition of "clinically meaningful improvement" may also differ in adults compared to infants, with a focus on maintaining existing abilities or slowing decline being highly valuable.
Table 2: Summary of Key Clinical Trial Results for Nusinersen (ENDEAR, CHERISH, NURTURE)
Trial Name (Population) | Primary Endpoint(s) | Key Efficacy Results (Nusinersen vs. Control/Natural History) | Key Long-Term Outcomes |
---|---|---|---|
ENDEAR (Infantile-Onset SMA, ≤7 months) | Motor milestone response (HINE); Event-free survival (death or permanent ventilation) | 51% motor milestone responders (Nusinersen) vs. 0% (sham); 47% reduction in risk of death/permanent ventilation. 71% CHOP INTEND responders (Nusinersen) vs. 3% (sham). 2 | SHINE extension: Maintained/improved motor function up to ~6 years. 28 |
CHERISH (Later-Onset SMA, ages 2-12, non-ambulatory) | Change in HFMSE score at 15 months | Mean HFMSE change: +3.9 points (Nusinersen) vs. -1.0 point (sham); treatment difference 4.9 points. 57% improved (Nusinersen) vs. 26% (sham). Improved HRQoL & reduced caregiver burden. 4 | SHINE extension: Maintained/improved HFMSE, ULM, 6MWT up to ~6 years. 28 |
NURTURE (Presymptomatic SMA, ≤6 weeks at first dose) | Prevention/delay of symptoms, respiratory intervention, death; Motor milestone achievement | ~5 years: All 25 alive, no permanent ventilation. 23/25 walking independently. Significant milestone achievement vs. natural history. HFMSE scores improved. 22 | Ongoing, extended to 8 years of age. Continued safety and efficacy. 22 |
Nusinersen has an established safety profile based on data from clinical trials and post-marketing experience. While generally considered favorable, particularly in the context of its significant benefits for SMA, there are known adverse reactions and risks that require careful monitoring.
The adverse reactions associated with Nusinersen can vary somewhat depending on the patient population (e.g., infantile-onset vs. later-onset SMA) and may also be related to the intrathecal administration procedure itself.
Common Adverse Reactions:
It is important to distinguish that many of the common side effects, particularly headache and back pain, are often directly related to the lumbar puncture procedure required for intrathecal administration, rather than being a direct pharmacological effect of Nusinersen itself.[4]
Serious Adverse Reactions (Rare):
While less common, some serious adverse events have been reported:
In the ENDEAR trial, no major serious safety concerns specifically related to the intrathecal injections were flagged, although it was noted that post-lumbar puncture syndrome is generally less common in children than in adults.[27] Despite the range of potential adverse events, the overall safety profile of Nusinersen is generally described as favorable and manageable, especially when considering the severity of untreated SMA and the transformative clinical benefits offered by the therapy.[4] Notably, in the CHERISH study, no children discontinued treatment due to adverse events.[29] Long-term follow-up studies, including NURTURE, SHINE, and adult observational studies, have not identified significant new safety signals.[22]
Table 3: Common and Serious Adverse Reactions Associated with Nusinersen
System Organ Class | Adverse Reaction | Frequency/Population Noted |
---|---|---|
Infections and Infestations | Lower respiratory infection | Common (Infantile-onset) 2 |
Upper respiratory infection | Common (General) 13 | |
Pneumonia | Common (General/Post-marketing) 5 | |
Aseptic meningitis | Rare/Serious 5 | |
Nervous System Disorders | Headache | Common (Later-onset, Adults, General) 2 |
Post-lumbar puncture syndrome | Common (General) 32 | |
Increased intracranial pressure | Rare/Serious 5 | |
Hydrocephalus | Rare/Serious (Post-marketing) 31 | |
Gastrointestinal Disorders | Constipation | Common (Infantile-onset) 2 |
Vomiting | Common (Later-onset, General) 2 | |
Nausea | Common (Adults, General) 5 | |
Musculoskeletal and Connective Tissue Disorders | Back pain | Common (Later-onset, Adults, General) 2 |
General Disorders and Administration Site Conditions | Pyrexia (Fever) | Common (Later-onset, General) 2 |
Immune System Disorders | Hypersensitivity reactions (e.g., rash, itching, swelling) | Rare/Serious 31 |
Metabolism and Nutrition Disorders | Hyponatremia (severe) | Very Rare/Serious (one infant in trial) 13 |
The use of Nusinersen is associated with certain warnings and precautions, primarily related to potential effects on coagulation and renal function. These are often considered potential class effects for antisense oligonucleotides (ASOs).
Thrombocytopenia and Coagulation Abnormalities:
There is an increased risk for bleeding complications associated with low platelet counts (thrombocytopenia) and other coagulation abnormalities (coagulopathy). This has been observed after the administration of some similar ASO therapies.2
Renal Toxicity:
An increased risk of kidney damage, including potentially fatal glomerulonephritis (inflammation of the kidney's filtering units), has also been observed after the administration of some ASO therapies.2 Nusinersen is known to be present in and excreted by the kidneys.8
The warnings for thrombocytopenia and renal toxicity appear to be partly based on observations with other ASOs, suggesting a potential class effect. However, data specific to Nusinersen, while still mandating careful monitoring, suggest that these risks may be manageable or less severe for this particular drug than initially extrapolated from the broader ASO class. The mandated laboratory monitoring before each dose is a critical risk mitigation strategy, allowing for early detection of potential adverse effects and timely intervention.
Post-Lumbar Puncture Syndrome:
Headache, back pain, and other symptoms consistent with post-lumbar puncture syndrome are common following the intrathecal administration procedure.32
Nusinersen (Spinraza®) is contraindicated in patients with known hypersensitivity to the active substance (nusinersen) or to any of the excipients contained in the formulation.[16] This is a standard contraindication for most pharmaceutical products, aimed at preventing severe allergic reactions in individuals with known sensitivities.
The potential for drug-drug interactions with Nusinersen appears to be relatively low, primarily due to its metabolism.
Nusinersen is not metabolized by the cytochrome P450 (CYP450) enzyme system in the liver; it is not a substrate for, nor an inhibitor or inducer of, these enzymes.5 This significantly reduces the likelihood of pharmacokinetic interactions with other drugs that are metabolized via CYP450 pathways, which is a common source of drug interactions. This characteristic is advantageous, as many SMA patients may require concomitant medications for other conditions.
Formal drug interaction studies with Nusinersen are limited. However, some drug interaction databases list potential interactions. For example, Drugs.com identifies 4 moderate drug interactions for nusinersen: dimethyl fumarate, diroximel fumarate, monomethyl fumarate, and remdesivir.[42] The specific mechanisms or detailed clinical significance of these listed interactions are not fully elaborated in the provided materials.
Disease interactions noted include bleeding risks and renal toxicity.[42] These are related to Nusinersen's own potential adverse effect profile and underscore the importance of the monitoring precautions discussed previously. Although not explicitly stated as direct interactions, caution would be pharmacologically prudent if co-administering Nusinersen with other drugs known to be nephrotoxic or to significantly affect coagulation, due to the potential for additive adverse effects.
Table 4: Known Drug and Disease Interactions with Nusinersen
Interacting Agent/Condition | Severity (Source: Drugs.com) | Potential Clinical Significance/Mechanism (Inferred or General) | Management Recommendations |
---|---|---|---|
Drugs: | |||
Dimethyl fumarate | Moderate 42 | Not specified; potential for overlapping adverse effects or monitoring needs. | Usually avoid combinations; use only under special circumstances. Consult prescriber. |
Diroximel fumarate | Moderate 42 | Not specified; potential for overlapping adverse effects or monitoring needs. | Usually avoid combinations; use only under special circumstances. Consult prescriber. |
Monomethyl fumarate | Moderate 42 | Not specified; potential for overlapping adverse effects or monitoring needs. | Usually avoid combinations; use only under special circumstances. Consult prescriber. |
Remdesivir | Moderate 42 | Not specified; potential for overlapping adverse effects (e.g., renal) or monitoring needs. | Usually avoid combinations; use only under special circumstances. Consult prescriber. |
Disease Conditions: | |||
Bleeding risks / Coagulopathy | Interaction noted 42 | Nusinersen has warnings for thrombocytopenia/coagulopathy. Pre-existing conditions may exacerbate risk. | Monitor platelet counts, PT, aPTT as per prescribing information. Caution. |
Renal toxicity / Renal Impairment | Interaction noted 42 | Nusinersen has warnings for renal toxicity and is renally excreted. Pre-existing impairment may increase risk or alter drug handling. | Monitor renal function (urine protein) as per prescribing information. Caution in patients with renal impairment. |
The recommended dosage of Nusinersen (Spinraza®) is 12 mg, administered as a single 5 mL intrathecal injection.[2] The dosing regimen consists of a series of loading doses followed by regular maintenance doses.
Loading Doses:
Treatment is initiated with four loading doses:
Maintenance Doses:
Following the completion of the loading dose series, a maintenance dose of 12 mg is administered once every 4 months.2 This 4-monthly interval is based on the long terminal elimination half-life of Nusinersen in the CSF.8
It is strongly recommended that Nusinersen treatment be initiated as early as possible after the diagnosis of SMA has been confirmed.[4] Clinical trial data have consistently shown that earlier intervention leads to better clinical outcomes.[2]
Duration of Treatment:
Information on the very long-term efficacy of Nusinersen is continually being gathered through ongoing studies and real-world experience. The need for continuation of therapy should be reviewed regularly by the treating physician and considered on an individual basis, depending on the patient's clinical presentation, response to therapy, and overall benefit-risk assessment.16 For many patients, treatment is anticipated to be lifelong.2
Table 5: Nusinersen (Spinraza®) Dosing Schedule
Dose Number | Timing | Dose |
---|---|---|
Loading Dose 1 | Day 0 (Initiation) | 12 mg (5 mL) |
Loading Dose 2 | Day 14 (2 weeks after Dose 1) | 12 mg (5 mL) |
Loading Dose 3 | Day 28 (2 weeks after Dose 2) | 12 mg (5 mL) |
Loading Dose 4 | Day 63 (approx. 9 weeks after Dose 3, or 30 days after Dose 3 per SmPC) | 12 mg (5 mL) |
Maintenance Doses | Every 4 months thereafter | 12 mg (5 mL) |
Note: Some variations in describing the interval for the 4th loading dose exist (e.g., "within first two months" [2], "9 weeks" [4], "Day 63" [16]). Clinicians should refer to the most current local prescribing information.
Nusinersen is administered exclusively by intrathecal injection via lumbar puncture, delivering the medication directly into the CSF.[2] This procedure must be performed by healthcare professionals who are experienced in conducting lumbar punctures, particularly in the SMA patient population which may present unique anatomical challenges.[4]
Due to the nature of the procedure and the patient population (which includes infants and young children), sedation or anesthesia may be required before administration to ensure patient comfort and procedural success.[4] The intrathecal route, while necessary for CNS drug delivery, can present challenges, especially with repeated administrations over a patient's lifetime. Conditions common in SMA, such as severe scoliosis (curvature of the spine) or previous spinal surgeries, can make accessing the intrathecal space difficult and may necessitate the use of imaging guidance (e.g., ultrasound or fluoroscopy) for accurate needle placement. These procedural complexities underscore the need for specialized clinical settings and expertise for the administration of Nusinersen.
The Summary of Product Characteristics (SmPC) provides specific guidance on managing missed or delayed maintenance doses of Nusinersen to help maintain therapeutic coverage.16
If a maintenance dose is delayed:
This guidance acknowledges the real-world challenges that can arise in adhering to a strict lifelong intrathecal therapy schedule, such as intercurrent illnesses, logistical issues, or patient/family circumstances. The recommendations aim to restore therapeutic drug levels while balancing practical considerations and safety, such as avoiding overly frequent lumbar punctures.
Nusinersen is indicated for the treatment of SMA in pediatric patients, encompassing infants, children, and adolescents.[2] The most robust clinical evidence for Nusinersen's efficacy and safety comes from studies conducted in this population. Pivotal trials such as ENDEAR (infantile-onset SMA), CHERISH (later-onset SMA), and NURTURE (presymptomatic SMA) have all demonstrated significant benefits in pediatric patients, as detailed in Section 4.[2] These trials have established Nusinersen as a foundational therapy for pediatric SMA, with the most profound effects observed when treatment is initiated early in the disease course or presymptomatically. The standard recommended dose of 12 mg is used across the pediatric age range.[2] While regulatory documents from Japan have mentioned potential dose adjustments for infants aged $\le$2 years due to smaller CSF volume, the globally accepted standard remains 12 mg per administration.[25]
The FDA approval for Nusinersen extends to adult patients with SMA.[2] However, the pivotal registration trials (ENDEAR, CHERISH, NURTURE) did not include adult participants.[32] The initial approval for adults was likely based on an understanding of SMA as a continuous disease spectrum from infancy through adulthood, the shared underlying pathophysiology, and the unmet need in this population. Since the initial approvals, real-world observational studies have become increasingly important in providing evidence for Nusinersen's use and effects in adults with SMA.[33] These studies, as discussed in Section 4.4, have generally shown stabilization of disease or modest improvements in motor function in a proportion of treated adults, which is significant given the typically progressive nature of SMA or eventual decline even in slower-progressing adult forms.
Regarding geriatric patients (typically defined as aged 65 and older), clinical studies of Nusinersen have not included a sufficient number of individuals in this age group to determine whether they respond differently from younger adult patients or to establish a specific safety profile for this cohort.[32] Treatment decisions in older adults would be based on individual clinical assessment, considering potential comorbidities and the overall benefit-risk profile.
Renal Impairment:
Nusinersen has not been formally studied in patients with pre-existing significant renal impairment.16 Therefore, its safety and efficacy in this specific population have not been established, and the European SmPC recommends close observation if used in such patients.16 Nusinersen and its metabolites are known to be excreted by the kidneys 8, and potential renal toxicity is a warning associated with ASO therapies (see Section 5.2). However, a recent study from Turkey investigating the effects of Nusinersen in SMA patients (without severe pre-existing renal disease) indicated that the treatment did not cause significant new renal dysfunction. Transient proteinuria was observed but was generally reversible.40 This suggests that while monitoring of renal function (via urine protein testing) is essential during Nusinersen therapy, the drug itself may not be directly nephrotoxic in individuals with normal baseline kidney function. The implications for patients with established severe renal impairment remain less clear due to the lack of specific studies.
Hepatic Impairment:
Similarly, Nusinersen has not been formally studied in patients with hepatic impairment.16 However, Nusinersen is not metabolized via the cytochrome P450 (CYP450) enzyme system in the liver; its metabolism occurs primarily via exonuclease activity in various tissues.8 Consequently, hepatic impairment is considered unlikely to significantly affect the pharmacokinetics of Nusinersen, and dose adjustments are generally not expected to be required for patients with liver dysfunction.16 While some general reports have mentioned instances of hepatic impairment in patients receiving Nusinersen, the causality and relationship to the underlying SMA or other factors remain unclear.37 The lack of hepatic CYP450 metabolism is a favorable characteristic, simplifying its use in patients who might have concurrent liver conditions.
Pregnancy:
There is limited information on the use of Nusinersen in pregnant women. Animal reproduction studies would typically inform potential risks, but detailed data from such studies are not extensively covered in the provided snippets. Due to the nature of the drug (an oligonucleotide with potential to affect rapidly developing systems) and the general lack of clinical trial data in pregnant women for ethical reasons, a precautionary approach is advised. Prescribing information generally states that Nusinersen may harm an unborn baby, and pregnancy should be avoided during treatment. If a patient becomes pregnant while receiving Nusinersen, they should inform their healthcare provider immediately.36 Pregnancy registries are often established for new drugs to collect post-marketing data on outcomes in women exposed during pregnancy, which helps to better understand any potential risks over time.43
Lactation:
Information regarding the presence of Nusinersen in human milk, the effects on the breastfed infant, or the effects on milk production is limited. Risk data concerning lactation is generally available, and patients are advised to inform their healthcare provider if they are breastfeeding or intend to breastfeed while on Nusinersen treatment.9 Decisions on whether to continue or discontinue breastfeeding during therapy should be made in consultation with a healthcare provider, weighing the benefits of breastfeeding against the potential risks to the infant and the importance of the medication to the mother.
No specific human data on teratogenicity for Nusinersen were found in the provided Japanese regulatory review documents.[19]
Nusinersen is a high-cost therapy, which has significant implications for healthcare systems and patient access. In the United States, the cost was reported in 2019 as US$750,000 for the first year of treatment (which includes more frequent loading doses) and US$375,000 annually thereafter for maintenance therapy.[3] More recent price guide information (likely from 2024/2025) indicates that the cost for a single 12 mg/5 mL vial of Spinraza® for intrathecal solution is approximately $156,187 for cash-paying customers.[45] Given that maintenance therapy involves administration every four months (i.e., three vials per year after the loading phase), this suggests an annual maintenance cost potentially higher than the 2019 figure, closer to $468,000 per year.
Pharmacoeconomic evaluations of Nusinersen have often concluded that, at its list price, the therapy exceeds conventional cost-effectiveness thresholds in many countries. Analyses have reported incremental cost-effectiveness ratios (ICERs) per quality-adjusted life year (QALY) gained ranging from approximately €464,891 to over €6.3 million for early-onset SMA, and from €493,756 to over €10.6 million for later-onset SMA, when compared to standard of care.[46] The Institute for Clinical and Economic Review (ICER) in the U.S. has also suggested that Spinraza® is overpriced relative to the clinical value it provides, estimating that its current price would need a substantial discount (approximately 10-fold) to meet a threshold of $100,000 per QALY gained, particularly for treating presymptomatic SMA Type 1.[47]
Despite these challenging pharmacoeconomic assessments, many countries have opted to reimburse Nusinersen.[46] This situation highlights a complex "value versus price" dilemma. Decision-makers often consider factors beyond traditional cost-effectiveness for orphan drugs that treat severe, life-threatening rare diseases with previously unmet medical needs. The profound clinical impact of Nusinersen, societal values, and equity considerations for rare disease populations appear to play a significant role in reimbursement decisions.[46]
It is also important to acknowledge the uncertainties inherent in economic models for therapies like Nusinersen. These include challenges in accurately quantifying utility values (a component of QALYs) in very young pediatric patients and the evolving understanding of long-term adherence to the demanding intrathecal treatment regimen and sustained real-world effectiveness, all of which can influence long-term cost-effectiveness projections.[46]
Recognizing the significant financial burden associated with Nusinersen, the manufacturer, Biogen, has established patient support programs aimed at facilitating access and mitigating out-of-pocket costs for eligible patients. In the United States, Biogen offers the SMA360° support program.[2] This program provides a range of services for patients and their families, designed to address nonmedical barriers to accessing Spinraza®. These services include assistance with navigating insurance coverage, facilitating the prior authorization process, providing information in the event of denied insurance claims, and connecting families with resources for financial assistance.[39]
A key component of these support initiatives is the Biogen Copay Program. This program is available for commercially insured patients who meet eligibility criteria and can help lower their out-of-pocket costs for Spinraza® to as low as $0. The copay assistance may potentially cover both the cost of the Spinraza® prescription and the treatment procedure itself. However, there may be annual caps on the amount of assistance a patient can receive, and eligibility can be affected by federal and state laws or other factors.[2] Patients covered by government healthcare programs like Medicare or Medicaid are typically not eligible for such copay programs.
Prior to full regulatory approval and widespread reimbursement, Expanded Access Programs (EAPs) or compassionate use programs were also made available by Biogen and Ionis Pharmaceuticals to provide Nusinersen to patients with infantile-onset SMA who had urgent medical needs.[26] These manufacturer-led support programs are essential for improving access to high-cost innovative therapies like Nusinersen, particularly within complex healthcare and insurance systems, by helping to reduce financial hurdles and navigate administrative complexities.
Nusinersen (Spinraza®) represents a landmark achievement in the treatment of spinal muscular atrophy. As the first FDA-approved therapy targeting the underlying molecular defect in SMA, it has fundamentally transformed the prognosis for individuals across a spectrum of disease types and ages. Clinical evidence has robustly demonstrated its ability to increase the production of functional SMN protein by modulating SMN2 splicing, leading to significant improvements in motor function, event-free survival (particularly in infantile-onset SMA), and overall quality of life. The benefits are most pronounced when treatment is initiated presymptomatically or very early in the disease course, as evidenced by the remarkable outcomes in the NURTURE trial. While the intrathecal administration route presents logistical challenges and the high cost of therapy poses significant economic and access hurdles, the safety profile of Nusinersen is generally considered manageable with appropriate monitoring for known potential risks such as thrombocytopenia and renal toxicity.
The therapeutic landscape for SMA is rapidly evolving. Nusinersen, while a pioneering agent, is now one of several approved disease-modifying therapies. Gene replacement therapy (e.g., onasemnogene abeparvovec - Zolgensma®) and other oral SMN2 splicing modifiers (e.g., risdiplam - Evrysdi®) offer alternative treatment modalities with different mechanisms of action, administration routes, and benefit-risk profiles.[44] The future role of Nusinersen will likely be shaped by ongoing long-term efficacy and safety data, comparative effectiveness research (though direct head-to-head trials are largely absent), patient and clinician preferences, and evolving cost-effectiveness considerations relative to these newer agents. Biogen's initiation of the DEVOTE trial, exploring higher doses of Nusinersen, indicates continued efforts to optimize its therapeutic potential.[28]
Ongoing long-term extension studies (such as SHINE and the NURTURE extension) and the collection of real-world evidence from adult patient registries will be critical for further delineating the durability of Nusinersen's effects, its very long-term safety profile (beyond 10-15 years), and its impact on non-motor aspects of SMA, which is a complex, multi-system disorder. Questions regarding the potential for waning efficacy over many years of treatment and the optimal management strategies for patients who may have a suboptimal response or who experience disease progression despite therapy will continue to be areas of active investigation.
Furthermore, the potential for combination or sequential therapies involving Nusinersen and other SMA treatments is an emerging area of research that may offer new avenues for maximizing therapeutic outcomes. Finally, persistent efforts from manufacturers, healthcare systems, patient advocacy groups, and policymakers will be crucial to address the ongoing challenges related to the high cost of Nusinersen and to ensure equitable global access for all individuals with SMA who could benefit from this life-altering therapy.
Published at: May 23, 2025
This report is continuously updated as new research emerges.