Medical Policy

Policy Num:      05.001.028
Policy Name:    Treatment for Spinal Muscular Atrophy
Policy ID:          [05.001.028]  [Ac / B / M+ / P+]  [5.01.28]


Last Review:       April 19, 2024
Next Review:      April 20, 2025

 

Related Policies: None

Treatment for Spinal Muscular Atrophy

Population Reference No.

Populations

Interventions

Comparators

Outcomes

1

Individuals:

    ·  Who are presymptomatic with a genetic diagnosis of spinal muscular atrophy and a minimum of 2 but less than 4 copies of SMN2

Interventions of interest are:

    ·         Nusinersen

Comparators of interest are:

    ·  Continued medical management

Relevant outcomes include:

    ·         Overall survival

    ·         Change in disease status

    ·         Functional outcomes

    ·         Quality of life

    ·         Treatment-related mortality

    ·         Treatment-related morbidity

2

Individuals:

    ·  With type I (infantile-onset) spinal muscular atrophy

Interventions of interest are:

     ·   Nusinersen

Comparators of interest are:

    ·  Continued medical management (respiratory, digestive, and orthopedic support)

Relevant outcomes include:

    ·         Overall survival

    ·         Change in disease status

    ·         Functional outcomes

    ·         Quality of life

    ·         Treatment-related mortality

    ·         Treatment-related morbidity

3

Individuals:

    ·   With types II or III spinal muscular atrophy

Interventions of interest are:

     ·   Nusinersen

Comparators of interest are:

    ·         Continued medical management (respiratory, digestive, and orthopedic support)

Relevant outcomes include:

    ·         Overall survival

    ·         Change in disease status

    ·         Functional outcomes

    ·         Quality of life

    ·         Treatment-related mortality

    ·         Treatment-related morbidity

4

Individuals:

     ·  With type I spinal muscular atrophy

Interventions of interest are:

     ·   Onasemnogene abeparvovec-xioi

Comparators of interest are:

    ·  Continued medical management (respiratory, digestive, and orthopedic support)

    ·   Nusinersen

Relevant outcomes include:

    ·         Overall survival

    ·         Change in disease status

    ·         Functional outcomes

    ·         Quality of life

    ·         Treatment-related mortality

    ·         Treatment-related morbidity

5

Individuals:

    ·  Who are presymptomatic with a genetic diagnosis of spinal muscular atrophy and less than 4 copies of SMN2

Interventions of interest are:

     ·   Onasemnogene abeparvovec-xioi

Comparators of interest are:

    ·   Nusinersen 

Relevant outcomes include:

    ·         Overall survival

    ·         Change in disease status

    ·         Functional outcomes

    ·         Quality of life

    ·         Treatment-related mortality

    ·         Treatment-related morbidity

6

Individuals:

    ·  With type I spinal muscular atrophy

Interventions of interest are:

    ·   Risdiplam

Comparators of interest are:

    ·   Continued medical management (respiratory, digestive, and orthopedic support)

    ·   Nusinersen

Relevant outcomes include:

    ·         Overall survival

    ·         Change in disease status

    ·         Functional outcomes

    ·         Quality of life

    ·         Treatment-related mortality

    ·         Treatment-related morbidity

7

Individuals:

    ·  With type II or III spinal muscular atrophy

Interventions of interest are:

     ·   Risdiplam

Comparators of interest are:

    ·  Continued medical management (respiratory, digestive, and orthopedic support)

    ·   Nusinersen

Relevant outcomes include:

    ·         Overall survival

    ·         Change in disease status

    ·         Functional outcomes

    ·         Quality of life

    ·         Treatment-related mortality

    ·         Treatment-related morbidity

Summary

Description

Spinal muscular atrophy is an inherited disorder caused by homozygous deletions or variants in the SMN1 gene. As a consequence of absent or low levels of survival motor neuron 1 protein, the motor neurons in the spinal cord degenerate, resulting in atrophy of the voluntary muscles of the limbs and trunk. Treatments in this review include 1) Nusinersen which is a synthetic antisense oligonucleotide designed to bind to a specific sequence in exon 7 of the SMN2 transcript causing the inclusion of exon 7 in the SMN2 transcript, leading to the production of full-length functional survival motor neuron 2 protein, which is very similar to SMN1. 2) Onasemnogene abeparvovec-xioi which is intended as a one-time gene replacement therapy designed to deliver a functional copy of the SMN1 gene to motor neuron cells of patients with spinal muscular atrophy. Because motor neurons are nondividing cells, it is postulated that once the SMN1 gene is incorporated in the cells, it would be retained over time and potentially allow for long-term, sustained survival motor neuron protein expression. 3) Risdiplam which is a once-daily self-administered oral therapy. It is a selective SMN2 splicing modifier designed to bind with specificity to SMN2 pre-mRNA to modulate SMN2 pre-mRNA splicing towards the production of full-length SMN2 mRNA.

Summary of Evidence

Presymptomatic Patients with a Genetic Diagnosis of Spinal Muscular Atrophy and a Minimum of 2 but Less Than 4 Copies of SMN2

For individuals who are presymptomatic with a genetic diagnosis of spinal muscular atrophy and a minimum of 2 but less than 4 copies of SMN2 who receive nusinersen, the evidence includes an open-label single-arm trial. Relevant outcomes are overall survival, change in disease status, morbid events, functional outcomes, quality of life, and treatment-related mortality and morbidity. After a median of 2.9 years of follow-up of 25 infants who received the first dose of nusinersen at a median age of 22 days and are now median age of 34.8 months, 100% were alive, 100% achieved the ability to sit without support, 92% achieved the ability to walk with assistance, and 88% achieved the ability to walk independently. While none required permanent ventilation or tracheostomy, 4 (16%) participants with 2 SMN2 copies utilized respiratory support for ≥6 h/day for ≥7 consecutive days that was initiated during acute, reversible illnesses. Results reported with additional 2 years of follow-up (data cut-off: February 15, 2021) demonstrated durability of treatment effect, and favorable safety profile after ~5 yeas of nusinersen treatment. These results demonstrate that early treatment resulted in the achievement of motor milestones among patients who are not likely to attain them without treatment. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Infantile-Onset or Type I Spinal Muscular Atrophy

For individuals who have type I (infantile-onset) spinal muscular atrophy who receive nusinersen, the evidence includes a randomized, double-blind, controlled trial. Relevant outcomes are overall survival, change in disease status, morbid events, functional outcomes, quality of life, and treatment-related mortality and morbidity. The phase 3 confirmatory ENDEAR trial (N =121) showed clinically meaningful and statistically significant improvement in motor milestones, event-free survival, and overall survival that exceeded those seen in the control group, with an acceptable safety profile. The proportion of patients, who met the primary endpoint responder definition of achieving motor milestones, was 51% in the nusinersen arm compared with 0% in the sham-controlled arm. Further, the hazard ratio for event-free survival was 0.53 favoring nusinersen over sham-controlled. It is notable, however, that 50% of nusinersen-treated subjects did not achieve the primary endpoint motor milestone response. Only a small proportion of patients (6%) gained the ability to sit without assistance. On average, the mean motor milestone score in nusinersen-treated patients improved by 3 points over 6 months. Given the limited data on the durability of response, long-term safety, and lack of efficacy in a substantial number of patients continued risk-benefit assessment of long-term treatment with nusinersen is necessary. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Type II and III Spinal Muscular Atrophy

For individuals who have type II or III spinal muscular atrophy who receive nusinersen, the evidence includes a double-blind, randomized controlled trial. Relevant outcomes are overall survival, change in disease status, morbid events, functional outcomes, quality of life, and treatment-related mortality and morbidity. Efficacy findings from single-arm studies of type II and III spinal muscular atrophy are difficult to interpret because these trials used a wide range of nusinersen doses and lacked control arms. The phase 3 confirmatory CHERISH trial (N =126) showed clinically meaningful and statistically significant improvement in motor milestones (measured using Hammersmith Functional Motor Scale–Expanded scores) that exceeded those seen in the control group (difference of 5.9 points favoring nusinersen over sham control, p<.001). The respective proportion of patients with clinically meaningful improvements in Hammersmith scores greater than 3 points was 57% vs 26% (p<.001). Multiple secondary endpoints also showed a consistency in treatment effect favoring nusinersen over sham control. Given the limited data on the durability of response, long-term safety, and lack of efficacy in a substantial number of patients continued risk-benefit assessment of long-term treatment with nusinersen is necessary. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Onasemnogene Abeparvovec-Xioi

Infantile-Onset or Spinal Muscular Atrophy Type I

For individuals who have spinal muscular atrophy type I (infantile-onset) who receive onasemnogene abeparvovec-xioi, the evidence includes 2 single-arm studies. Relevant outcomes are overall survival, change in disease status, functional outcomes, quality of life, and treatment-related mortality and morbidity. The FDA approval was based on a pooled analysis of 21 patients from the 2 single-arm studies. The observed treatment effect on survival, event-free survival, and achievement of motor functions is beyond what is typical based on the known natural history of patients with spinal muscular atrophy type I with 2 copies of SMN2. Results of the phase 3 confirmatory study (STRIVE-US) published after the FDA approval were consistent with previously available findings at the time of approval. Results of an ongoing study to assess long-term safety and durability of response in infants with SMA type 1 with a median time since dosing of 5.2 years showed that the developmental milestones achieved in the phase 1 clinical trial were maintained and new milestones gained. Thirteen of 15 original patients were included in the analysis. All 10 patients in the therapeutic-dose cohort remained alive and without the need for permanent ventilation. All 10 patients treated with the therapeutic dose maintained previously acquired motor milestones. Two patients attained the new milestone of “standing with assistance” without the use of nusinersen. However, 7 of the 13 subsequently received concomitant nusinersen. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Presymptomatic Patients with a Diagnosis of Spinal Muscular Atrophy and Less Than 4 Copies of SMN2

For individuals who are presymptomatic with a genetic diagnosis of spinal muscular atrophy and less than 4 copies of SMN2 who receive onasemnogene abeparvovec-xioi, the evidence includes one single-arm study- SPR1NT. Relevant outcomes are overall survival, change in disease status, functional outcomes, quality of life, and treatment-related mortality and morbidity. The SPR1NT trial included infants less than 6 weeks of age who did not display any symptoms of SMA at the time of infusion. The trial was divided into 2 cohorts based on SMN2 copy number. All patients irrespective of SMN2 copy numbers achieved their respective primary endpoint, able to sit without support for at least 30 seconds at 18 months of age for 2 copies of SMN2 or ability to stand for 3 seconds or more without assistance at 24 months for those with 3 copies of SMN2. Multiple secondary endpoints were also supportive of clinical benefit including achievement of age-appropriate gross motor milestones and functions and independence from nutritional and respiratory support. In the natural history of SMA Type 1, untreated children with 2 or 3 copies of the SMN2 backup gene would not achieve such skills. These results demonstrate that early treatment resulted in the achievement of motor milestones among patients who are not likely to attain them without treatment. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Risdiplam

Spinal Muscular Atrophy Type I

For individuals who have symptomatic SMA type I (infantile-onset) who receive risdiplam, the evidence includes 1 single-arm prospective phase 2/3 FIREFISH study. Relevant outcomes are overall survival, change in disease status, functional outcomes, health status measures, quality of life, and treatment-related mortality and morbidity. Results of a part 1 study (dose-finding and exploratory) in which 4 patients received low dose and 17 patients received high dose risdiplam are available. The results of the part 1 study (dose-finding and exploratory) showed 41% (7 of 17 infants) were able to sit without support for at least 5 seconds as assessed by BSID-III after 12 months of treatment. After a minimum follow-up of 12 and 23 months of treatment with risdiplam, 90% (19/21) and 81% (17/21) of patients were alive without permanent ventilation. Results of a part 2 study in which 41 patients received the FDA recommended dose reported that after 12 months of treatment, 29% were able to sit without support for at least 5 seconds, 56% achieved a CHOP-INTEND score of 40 or higher, 90% achieved an increase of at least 4 points from baseline in the CHOP-INTEND score, 78% achieved a HINE-2 motor milestone response, and 85% survived without permanent ventilation. Results of part 2 of the study in which 41 patients received FDA recommended dose reported that after 12 and 24 months of treatment, 29% and 61% were able to sit without support for at least 5 seconds respectively, 56% and 76% achieved a CHOP-INTEND score of 40 or higher respectively and 85% and 83% survived without permanent ventilation respectively. The observed treatment effect on motor functions and event-free survival is beyond what is typical based on the known natural history of patients with SMA type I with 2 copies of SMN2. However, there is limited data to assess the long-term durability of treatment effect as well as safety-related to adverse events that are rare or have delayed onset. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Spinal Muscular Atrophy Type II and III

For individuals who have symptomatic type II or III SMA who receive risdiplam, the evidence includes a single ongoing double-blind, 2-part, placebo-controlled (SUNFISH) study. Relevant outcomes are overall survival, change in disease status, functional outcomes, health status measures, quality of life, and treatment-related mortality and morbidity. Pediatric or adult patients (2-25 years old) with type II or III SMA patients were enrolled in the SUNFISH study. Results of 180 patients from part 2 with 12 months follow-up reported least-square mean change in MFM-32 from baseline was greater in patients receiving risdiplam compared with placebo with a difference of 1.55 (95% CI: 0.30 to 2.81). Responder analysis (using a threshold of increase in 3 or more points in MFM-32 as being clinically meaningful) showed that 38% of those receiving risdiplam met the criteria of achieving a clinically meaningful improvement in motor function compared to 24%. Upper limb function measured by RULM showed a 1.6 point improvement in the risdiplam-treated group compared to no change in the placebo group. At month 24 of risdiplam treatment, 32% of patients demonstrated improvement (a change of ≥3) from baseline in MFM32 total score; 58% showed stabilization (a change of ≥0). Overall, gains in motor function at month 12 were maintained or improved upon at month 24. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Additional Information

Not applicable.

Objective

The objective of this evidence review is to assess whether treatment of spinal muscular atrophy improves the net health outcome.

Policy Statements

Nusinersen

Initial Treatment

Nusinersen may be considered medically necessary if all the following conditions are met:

  1. Diagnosis of spinal muscular atrophy confirmed by genetic testing demonstrating bi-allelic mutations in the survival motor neuron 1 (SMN1) gene as stated below:
      1. Deletion of both copies of the SMN1 gene OR

      2. Compound heterozygous mutations of the SMN1 gene (defined below):

        1. Pathogenic variant(s) in both copies of the SMN1 gene

        2. Pathogenic variant in 1 copy and deletion of the second copy of the SMN1 gene.

  2. If the individual is symptomatic, documentation of a genetic test confirms 2, 3, or 4 copies of the SMN2 gene; OR if the individual is asymptomatic, documentation of a genetic test confirms 2 or 3 copies of the SMN2 gene.

  3. The individual is not on permanent ventilator dependence.
  4. Medication is being administered at the United States Food and Drug Administration approved dosing by a healthcare professional (see Policy Guidelines).

  5. The individual is not concurrently enrolled in a clinical trial for any experimental therapy for spinal muscular atrophy.

  6. Prescribed by a neurologist with expertise in treating spinal muscular atrophy.

Initial authorization period is for 12 months.

Continuation of Treatment

Incremental reauthorization for nusinersen may be considered medically necessary if the following conditions are met:

  1. The individual continues to meet initial treatment criteria cited above;

  2. Documented evidence to support clinically meaningful improvement in motor milestones during the previous treatment period. Obtain a baseline motor milestone score from a validated assessment such as Hammersmith Infant Neurologic Exam (HINE), Children’s Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP-INTEND), etc.

Reauthorization period is for 12 months.

Nusinersen is considered investigational for all other indications.

Concurrent use of nusinersen with onasemnogene abeparvovec-xioi and/or risdiplam is considered investigational.

Use of nusinersen after administration of onasemnogene abeparvovec-xioi is considered investigational.

Onasemnogene Abeparvovec-Xioi

Onasemnogene abeparvovec-xioi may be considered medically necessary if all of the following conditions are met:

  1. Diagnosis of spinal muscular atrophy confirmed by genetic testing demonstrating bi-allelic mutations in the survival motor neuron 1 (SMN1) gene as stated below:

    1. Deletion of both copies of the SMN1 gene OR

    2. Compound heterozygous mutations of the SMN1 gene (defined below):

      1. Pathogenic variant(s) in both copies of the SMN1 gene OR

      2. Pathogenic variant in 1 copy and deletion of the second copy of the SMN1 gene.

  2. Documentation of a genetic test confirms no more than 3 copies of the SMN2 gene.
  3. The individual is less than 2 years of age at the time of infusion of onasemnogene abeparvovec-xioi.

  4. Documentation of baseline laboratory assessments such as AST, ALT, total bilirubin, and prothrombin time.
  5. The individual does not have advanced spinal muscular atrophy (e.g., complete paralysis of limbs, permanent ventilator dependence).

  6. Baseline anti-adeno-associated virus serotype 9 (AAV9) antibody titers < 1:50.

  7. Prescribed by a neurologist with expertise in treating spinal muscular atrophy.

Repeat treatment or ante-partum use of onasemnogene abeparvovec-xioi is considered investigational.

Onasemnogene abeparvovec-xioi is considered investigational for all other indications.

Concurrent use of onasemnogene abeparvovec-xioi with nusinersen and/or risdiplam is considered investigational.

Use of nusinersen and/or risdiplam after administration of onasemnogene abeparvovec-xioi is considered investigational.

Risdiplam

Initial Treatment

Risdiplam may be considered medically necessary if all the following conditions are met:

  1. Diagnosis of spinal muscular atrophy confirmed by genetic testing demonstrating bi-allelic mutations in the survival motor neuron 1 (SMN1) gene as stated below:

    1. Deletion of both copies of the SMN1 gene OR

    2. Identification of pathogenic variant(s) in both copies of the SMN1 gene.

  2. If the individual is symptomatic, documentation of a genetic test confirms 2, 3, or 4 copies of the SMN2 gene; OR if the individual is asymptomatic, documentation of a genetic test confirms 2 or 3 copies of the SMN2 gene.

  3. Individual age is 25 years or less.

  4. The individual is not on permanent ventilator dependence.

  5. Medication is being administered at the United States Food and Drug Administration-approved dosing (see Policy Guidelines).
  6. The individual is not concurrently enrolled in a clinical trial for any experimental therapy for spinal muscular atrophy.

  7. Prescribed by a neurologist with expertise in treating spinal muscular atrophy.

Initial authorization period is for 12 months.

Continuation of Treatment

Incremental reauthorization for risdiplam may be considered medically necessary if the following conditions are met:

  1. The individual continues to meet initial treatment criteria cited above;

  2. Documented evidence to support clinically meaningful stabilization or improvement in motor milestones during the previous treatment period. Obtain a baseline motor milestone score from a validated assessment such as Hammersmith Infant Neurologic Exam (HINE), Children’s Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP-INTEND), etc.

Reauthorization period is for 12 months.

Risdiplam is considered investigational for all other indications.

Concurrent use of risdiplam with onasemnogene abeparvovec-xioi and/or nusinersen is considered investigational.

Use of risdiplam after administration of onasemnogene abeparvovec-xioi is considered investigational.

Policy Guidelines

Nusinersen

The recommended dose of nusinersen is 12 mg (5 mL) administered intrathecally by, or under the direction of, healthcare professionals experienced in performing lumbar punctures. Treatment is initiated with 4 loading doses; the first 3 loading doses should be administered at 14-day intervals while the fourth loading dose is administered 30 days after the third loading dose. Maintenance doses should be administered once every 4 months thereafter.

Dosing Limits

Initial: 4 loading doses
Maintenance: 1 dose every 4 months

Onasemnogene Abeparvovec-Xioi

The recommended dosage of onasemnogene abeparvovec-xioi is 1.1 × 1014 vector genomes (vg) per kg of body weight. It should be administered as an intravenous infusion over 60 minutes. Systemic corticosteroids equivalent to oral prednisolone at 1 mg/kg should be administered according to the U.S. Food and Drug Administration (FDA) approved prescribing label.

FDA has issued a black-box warning for onasemnogene abeparvovec-xioi due to the risk of acute serious liver injury and elevated aminotransferases. Individuals with pre-existing liver impairment may be at higher risk.

The FDA label states that “The safety and efficacy of ZOLGENSMA in individuals with anti-AAV9 antibody titers above 1:50 have not been evaluated.” Baseline anti-AAV9 antibody testing is performed prior to infusion using. Retesting may be performed if anti-AAV9 antibody titers are reported as >1:50.

Liver function (clinical exam, AST, ALT, total bilirubin, prothrombin time), platelet counts, and troponin-I levels should be monitored as per the prescribing label.

Where feasible, the individual's vaccination schedule should be adjusted to accommodate concomitant corticosteroid administration prior to and following onasemnogene abeparvovec-xioi infusion.

Use of onasemnogene abeparvovec-xioi in premature neonates before reaching full-term gestational age may not be recommended because concomitant treatment with corticosteroids may adversely affect neurological development.

Efficacy of onasemnogene abeparvovec-xioi in individuals with c.859G>C variant in SMN2 gene has not been evaluated.

A pre-symptomatic individual may be identified through the SMA newborn screening process and eligible for treatment. The requirements within the policy must be satisfied regardless of the SMA newborn screening.

Dosing Limits

1 injection per lifetime

Risdiplam

Risdiplam is administered orally once daily after a meal using a provided oral syringe. The recommended daily dosing of risdiplam is based on age and weight as follows:

Risdiplam should be avoided in individuals with hepatic impairment. It is recommended that individuals who can become pregnant undergo pregnancy testing prior to initiating treatment with risdiplam.

Dosing Limits

7 bottles per 84 days

SMN2 Copy Numbers

For detail refer to Table 2 in the “Background” Section. Generally, the number of SMN2 gene copies is inversely related to the severity of spinal muscular atrophy. Higher numbers typically correlate with less severe disease. Among individuals clinically diagnosed with type I spinal muscular atrophy, 73% had 2 copies of SMN2, type II spinal muscular atrophy individuals 82% had 3 copies of SMN2, and type III spinal muscular atrophy individuals 51% had 3 and 46% had 4 copies of SMN2. Conversely, the probability that an unaffected child who has been tested after birth and has been found to carry a homozygous SMN1 deletion, the probability of developing type I spinal muscular atrophy if the child has 1 or 2 copies of SMN2 is greater than 97%. Those with 3 copies of SMN2 have an 83% probability of developing type II spinal muscular atrophy and those with 4 copies of SMN2 have an 84% probability of developing type III spinal muscular atrophy.

Nusinersen, onasemnogene abeparvovec-xioi, and risdiplam have not been evaluated in individuals with Type 0 or Type IV SMA.

Coding

See the Codes table for details.

Benefit Application

Benefits are determined by the group contract, member benefit booklet, and/or individual subscriber certificate in effect at the time services were rendered.  Benefit products or negotiated coverages may have all or some of the services discussed in this medical policy excluded from their coverage.

BlueCard/National Account Issues

State or federal mandates (eg, Federal Employee Program) may dictate that certain U.S. Food and Drug Administration-approved devices, drugs, or biologics may not be considered investigational, and thus these devices may be assessed only by their medical necessity.

Background

Spinal Muscular Atrophy

Spinal muscular atrophy is a rare autosomal recessive genetic disorder caused by homozygous deletions or variants in the SMN1 gene located on chromosome 5. This gene produces the “survival of motor neuron” protein (SMN1), which is essential for motor neuron functioning. In 95% of cases of spinal muscular atrophy, there is a homozygous deletion of exon 7 in the SMN1 gene. The remaining 5% of cases are compound heterozygotes for SMN1 exon 7 deletions and small intragenic variants.1, Due to absent or low levels of the survival motor neuron 1 protein, motor neurons in the spinal cord degenerate, resulting in atrophy of the voluntary muscles of the limbs and trunk affecting the ability to crawl, walk, sit up, and control head. In more severe cases, feeding, swallowing, and breathing are affected as well. The exact role of the survival motor neuron protein in motor neurons has not been completely elucidated, and levels of the survival motor neuron protein required for optimal functioning are unknown.2,

There is wide phenotypic heterogeneity in spinal muscular atrophy, as summarized in Table 1. This is due to the presence of SMN2, a modifying/backup gene, also located on chromosome 5, which is 99% identical to SMN1. However, 70% to 90% of the SMN2 compensatory protein produced by this gene is defective and unstable due to the lack of exon 7.3, The number of copies of the SMN2 gene varies widely (range, 0-6), resulting in a less severe form of spinal muscular atrophy among those with more copies of the SMN2 gene and vice-versa.4, The relation between the SMN2 copy number and spinal muscular atrophy phenotype is summarized in Table 2. These data were generated from DNA samples of 375 patients with spinal muscular atrophy who previously had been classified as follows: 188 with spinal muscular atrophy type I, 110 with spinal muscular atrophy type II, and 77 with spinal muscular atrophy type III.5,

Table 1. Characteristics and Subtypes of Spinal Muscular Atrophy
 
Type of SMA Age at Symptoms Onset Life Span Highest Motor Milestone Achieved SMN2 Copy Numbera
Type 0 (antenatal-onset SMA) Prenatal <6 months Little ability to move and may be unable to breathe and swallow independently. 1
Type I (infantile SMA or Werdnig-Hoffman disease) 0-6 months <2 y without respiratory support Never rolls or sits unsupported. 2
Type II (intermediate SMA or Dubowitz disease) <18 months >2 y; »70% alive at 25 years of age Sits independently once properly positioned; sometimes stands but never able to walk. 3 or 4
Type III (Kugelberg-Welander disease)        
Subtype IIIa >18 months to 3 years Similar to that of the general population Sits, stands, and walks independently until puberty; many no longer walk after puberty. Never runs or jumps well. 3 or 4
Subtype IIIb >3 years Similar to that of the general population Sits, stands, and walks independently until puberty; many no longer walk after puberty. Walks, runs, jumps, and can participate in sports. 4
Type IV (adult-onset SMA) >21 years Similar to that of the general population Similar to that of the general population. 4-8
Adapted from the Muscular Dystrophy Association (n.d.),6, National Organization for Rare Disorders (2012),7,Zerres et al (1995),8, Finkel et al (2014),9, and Rudnik-Schoneborn et al (2001).10,SMA: spinal muscular atrophy.a Quantitative analysis of SMN2 copies in 375 patients showed that 80% of SMA type I carry 1 or 2 SMN2 copies, 82% with SMA type II carry 3 SMN2 copies, and 96% with SMA type III carry 3 or 4 SMN2 copies.5,Among 113 patients with SMA type I, 9 with 1 SMN2 copy lived <11 months, 88 of 94 with 2 SMN2 copies lived <21 months, and 8 of 10 with 3 SMN2 copies lived 33 to 66 months.11,
Table 2. Relation Between SMN2 Copy Numbers and Spinal Muscular Atrophy Phenotype
Type of SMA Percent With 1
SMN2 Copy
Percent With 2 SMN2 Copies Percent With 3 SMN2 Copies Percent With 4 SMN2 Copies
Type I 6.9 73.4 19.7 0
Type II 0 10.9 81.8 7.3
Type III 0 3.9 50.6 45.5
  Probabilitya of SMA Type I Probabilitya of SMA Type II Probabilitya of SMA Type III  
1 SMN2 copy 99.9 0 0  
2 SMN2 copies 97.3 2.7 0  
2 SMN2 copies 7.2 82.8 10.0  
4 SMN2 copies 1.6 14.8 83.6  
Adapted from Feldkotter et al (2002)5,SMA: spinal muscular atrophy.a Probability that an unaffected child who has been tested after birth and has been found to carry a homozygous SMN1 deletion will develop SMA type.

Diagnosis

Spinal muscular atrophy can be diagnosed using multiple molecular genetic testing techniques such as multiplex ligation-dependent probe amplification or quantitative polymerase chain reaction or a comprehensive next-generation sequencing-based approach. Individuals are classified as having spinal muscular atrophy if they have a homozygous deletion of the SMN1 gene or a homozygous absence of the SMN1 gene due to gene conversion (ie, SMN1 gene conversion to SMN2 gene) or a compound heterozygote variant in the SMN1 gene. Individuals are defined as carriers if they have 1 copy of the SMN1 gene on 1 chromosome and no copies on the other or 2 copies of the SMN1 gene on 1 chromosome and no copies on the other. Assessing SMN2 copy numbers as part of a diagnostic workup is important because it can provide critical information on disease progression and assist in possible clinical trial enrollment or treatment.

Because spinal muscular atrophy symptom onset may occur shortly after birth to months to years later, estimating the incidence and prevalence of spinal muscular atrophy subtypes is difficult. The incidence, as reported in the literature, is more precisely a birth prevalence rate, which is estimated between 9.1 and 10 per 100,000 live births,12,13, which translates to 500 new spinal muscular atrophy cases annually.

Treatment

Medical management of spinal muscular atrophy patients includes respiratory, nutritional, and musculoskeletal supportive care. Respiratory management includes airway clearance, antibiotic treatment of infections, noninvasive and invasive ventilation. Nutritional management includes changing food consistency, gastrostomy tube feeding, and dietician assessment. Musculoskeletal supportive care includes a variety of interventions such as equipment for mobility, teaching self-care and function, physiotherapy, spinal surgery, posture and pain management, regular exercise, and scoliosis surgery. The type and extent of supportive care can affect survival in infant-onset disease (eg, gastrostomy feeding and noninvasive/invasive ventilation).

Nusinersen is a modified antisense oligonucleotide (a synthetic genetic material) that binds to a specific sequence in the intron downstream of exon 7 of the SMN2 transcript; nusinersen causes the inclusion of exon 7 in the SMN2 transcript, leading to the production of full length functional SMN2 protein.14,

Risdiplam is a SMN2 splicing modifier that increases exon 7 inclusion in SMN2 messenger ribonucleic acid (mRNA) transcripts and the production of full-length SMN protein in the brain.15,

Onasemnogene abeparvovec-xioi, a 1-time gene replacement therapy is intended as an intravenous infusion for patients with spinal muscular atrophy type I and an intrathecal infusion for spinal muscular atrophy type II.

Regulatory Status

On December 23, 2016, nusinersen (Spinraza™; Biogen) was approved by the U.S. Food and Drug Administration (FDA) for the treatment of pediatric and adult patients with spinal muscular atrophy.

On May 24, 2019, onasemnogene abeparvovec-xioi (Zolgensma®; Avexis) was approved by the FDA for the treatment of pediatric patients less than 2 years of age with spinal muscular atrophy with bi-allelic mutations in the survival motor neuron 1 gene.

On August 7, 2020, risdiplam (Evrysdi™; Genentech) was approved by the FDA for the treatment of spinal muscular atrophy in patients 2 months of age and older. On May 31, 2022, FDA approved a label extension for oral risdiplam to include infants younger than 2 months old with spinal muscular atrophy.

Rationale

This evidence review was created in February 2017 and has been updated regularly with searches of the PubMed database. The most recent literature update was performed through January 8, 2024.

Evidence reviews assess the clinical evidence to determine whether the use of technology improves the net health outcome. Broadly defined, health outcomes are the length of life, quality of life, and ability to function, including benefits and harms. Every clinical condition has specific outcomes that are important to patients and managing the course of that condition. Validated outcome measures are necessary to ascertain whether a condition improves or worsens; and whether the magnitude of that change is clinically significant. The net health outcome is a balance of benefits and harms.

To assess whether the evidence is sufficient to draw conclusions about the net health outcome of technology, 2 domains are examined: the relevance, and quality and credibility. To be relevant, studies must represent 1 or more intended clinical use of the technology in the intended population and compare an effective and appropriate alternative at a comparable intensity. For some conditions, the alternative will be supportive care or surveillance. The quality and credibility of the evidence depend on study design and conduct, minimizing bias and confounding that can generate incorrect findings. The randomized controlled trial (RCT) is preferred to assess efficacy; however, in some circumstances, non-randomized studies may be adequate. RCTs are rarely large enough or long enough to capture less common adverse events and long-term effects. Other types of studies can be used for these purposes and to assess generalizability to broader clinical populations and settings of clinical practice.

Promotion of greater diversity and inclusion in clinical research of historically marginalized groups (e.g., People of Color [African-American, Asian, Black, Latino and Native American]; LGBTQIA (Lesbian, Gay, Bisexual, Transgender, Queer, Intersex, Asexual); Women; and People with Disabilities [Physical and Invisible]) allows policy populations to be more reflective of and findings more applicable to our diverse members. While we also strive to use inclusive language related to these groups in our policies, use of gender-specific nouns (e.g., women, men, sisters, etc.) will continue when reflective of language used in publications describing study populations.

Nusinersen

Clinical Context and Therapy Purpose

The purpose of nusinersen in pediatric and adult patients who have spinal muscular atrophy is to provide a treatment option that is an improvement on existing therapies. Potential benefits of this therapy may include the following:

The following PICO was used to select literature to inform this review.

Populations

The relevant population of interest is individuals with a genetic diagnosis of spinal muscular atrophy.

Interventions

The therapy being considered is nusinersen.

Comparators

Prior to the availability of nusinersen, there was no U.S. Food and Drug Administration (FDA) approved treatment for spinal muscular atrophy. Medical management includes respiratory, nutritional, and musculoskeletal supportive care.

Outcomes

The general outcomes of interest are survival, functional ability, quality of life, and treatment-related mortality and morbidity. Because spinal muscular atrophy is a heterogeneous disease, measuring the impact of the intervention depends on the subtype of spinal muscular atrophy. For example, in infantile-onset spinal muscular atrophy (type I), comparing the achievement of motor milestones with the known natural history of spinal muscular atrophy is relevant, but the same may not be applicable for patients with late-onset spinal muscular atrophy (type III) in whom normal motor milestones may be delayed but nevertheless achieved or achieved but lost later. Age- and ability-appropriate motor function scales as they relate to the natural progression of spinal muscular atrophy are summarized in Table 3.

Table 3. Health Outcome Measures Relevant to Spinal Muscular Atrophy
Outcome Age Description Relevance
BSID-III
  • Appropriate for ages 1 to 42 months16,
  • Designed to measure movements of the limbs and torso in infants and toddlers
  • A range of motor functions including rolling, sitting upright, crawling, stepping motions, standing, walking, climbing stairs, running, maintaining balance, and other activities requiring full-body control or coordination16,
  • In the natural history of the disease, infants with Type 1 SMA are not expected to achieve independent sitting or any of the other subsequent milestones evaluated
CHOP INTEND
  • Appropriate for 3.8 mo to >4 y
  • Motor skills include 16 items scored on a scale of 0 (no response) to 4 (complete response) and total score ranges from 0 to 6417,18,
  • Score >40 rare in SMA type I with 2 SMN2 gene copies19,
  • Mean CHOP INTEND score in healthy infants (n=14; age, 3.3 mo) was 501.1 vs 20.2 in SMA type I (n=16; age, 3.7 mo)
HFMSE
  • Appropriate for individuals with SMA types II and III20,
  • Motor function includes 33 items from the Gross Motor Function Measure related to lying/rolling, crawling, crawling/kneeling, standing, and walking/running/jumping that are scored on a scale of 0 to 2, with a total score that ranges from 0 to 66, where lower scores indicate poorer motor function. On average, it can be conducted in 12 min.
  • Multiple studies have shown that HFMSE scores decline progressively in patients with SMA type II or III. However, there is conflicting data on whether such declines are linear.21,22,
HINE Section 2
  • Appropriate for infants 2-24 mo
  • Motor milestones include 8 items scored on a 5-point scale with 0 as the absence of activity, and a maximum score of 4 points
  • Infants with the most severe symptoms of SMA (early onset) may show a score of 0 on all 8 items of the HINE Section 223,
MFM-32
  • Appropriate for ages: 2 to 62 years
  • Evaluates gross and fine motor function in a broad patient population (weak nonambulant to stronger ambulant, and all levels of severity)
  • 3 domains including standing and transfers, axial limb and proximal limb motor function, and distal motor function
  • Scored using 4-point Likert scale based on subject’s maximal abilities without assistance (0 to 3)
  • Results of an observational retrospective analysis of 12 type 2 and 19 type 3 SMA patients showed a slow deterioration (-0.9 points/y and -0.6 points/y respectively).24,
  • Studies assessing the validity and reliability of MFM-32 support its use in longitudinal research in individuals with type 2 and non-ambulant type 3 SMA.25,26,
  • Results of a retrospective analysis of 81 patients with type 2 and non-ambulatory type 3 SMA aged 2–25 years suggest that a within-patient change of 3 to 4 points was meaningful. Further, the authors conclude that given the progressive nature of SMA, a 3 to 4-point improvement should not be viewed as minimal as arguably stabilization is also meaningful.27,
RULM
  • Appropriate for children and adults with SMA age ≥ 30 months
  • Evaluates upper limb function
  • Scale consists of 20 items that test proximal and distal motor
    functions of the arm. The total score ranges from 0, if all the items cannot be performed, to 37, if all the activities are achieved fully without any compensatory maneuvers.
  • Higher scores indicate higher function28,
  • The purpose of an upper limb scale for use in SMA is to assess change that occurs in motor performance of the upper limb overtime. Literature search did not identify threshold for clinically meaningful difference.
Natural history with and without SMA   Infants without SMA at 1 y29,:
  • 90% able to maintain head control, turn in sitting position (pivot), form a pincer grasp, play with feet, roll from prone to supine (and back), crawl on hands and knees
  • 79% able to stand unaided
  • 51% able to walk
At 18 mo:
  • 90% stand/walk unaided
Event-free survival rates in infants with SMA type I9,30,:
  • 50% by 8-10.5 mo
  • 25% by 13.6 mo
  • 8% by 20 mo
  • With the availability of nusinersen, conducting placebo-controlled trials in patients with SMA type I who face near-term mortality would be unethical. Therefore, good quality natural history data from SMA and non-SMA populations using validated cohorts are essential to assess relative health benefit over short- and long-term.
BSID: Bayley Scales of Infant and Toddler Development (Third Edition); CHOP INTEND: Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders; HFMSE: Hammersmith Functional Motor Scale-Expanded; HINE: Hammersmith Infant Neurological Examination; MFM-32: Motor Function Measure (32 items); RULM: Revised Upper Limb Module; SMA: spinal muscular atrophy.

Given the heterogeneity and varying life expectancies among patients with different spinal muscular atrophy subtypes, the timing of follow-up of studies to reasonably assess whether nusinersen offers a net health benefit will differ by spinal muscular atrophy subtypes as well as by the timing of treatment initiation relative to symptom onset. Given the significant uncertainty about the durability of the long-term benefits and safety of therapies, long-term data in an observational setting are also a requirement. The timing of outcomes measures relevant to spinal muscular atrophy subtypes is summarized in Table 4.

Table 4. Timing of Outcome Measures Relevant to Spinal Muscular Atrophy
SMA Subtype Purpose Timing
Presymptomatic with a genetic diagnosis of SMA and less than 4 copies of SMN2
  • To assess short-term benefit (efficacy & safety)
  • 6 mo to 1 y may be sufficient
Types I to III
  • To assess short-term benefit (efficacy and safety)
  • 1-2 y may be sufficient
Types I to III
  • To assess durability of benefit and delayed/rare adverse events
  • 10-15 y (survival, comparative development milestones vs natural history of SMA and non-SMA patients, safety)
SMA: spinal muscular atrophy.

Study Selection Criteria

Methodologically credible studies were selected using the following principles:

Population Reference No. 1 

Presymptomatic Patients with a Genetic Diagnosis of Spinal Muscular Atrophy and a Minimum of 2 But Less Than 4 copies of SMN2

Review of Evidence

Non-Randomized Studies

In the pivotal single arm NURTURE trial, ), 25 infants received nusinersen. Of the 25 infants, 15 had 2 copies of SMN2 (most likely to develop type 1 spinal muscular atrophy) and 10 had 3 copies of SMN2 (most likely to develop type II spinal muscular atrophy). Infants’ age at first dose ranged from 8 to 41 days (≤14 days, n=9; >14 to ≤28 days, n=12; >28 days, n=4). See Table 5 for study key characteristics summary.

Results are summarized in Table 6. The primary endpoint was time to death or respiratory intervention (invasive or non-invasive for ≥6 h per day continuously for ≥7 days or tracheostomy). The median (range) age at the first dose of nusinersen among enrolled infants was 22.0 (3–42) days. At the last visit, the median age was 34.8 months (range 25.7 to 45.4) which is generally considered past the expected age of symptom onset for spinal muscular atrophy Types I or II based on known natural history. All 25 children were alive and none required tracheostomy or permanent ventilation. Four (16%) participants with 2 SMN2 copies utilized respiratory support for ≥6 h/day for ≥7 consecutive days that was initiated during acute, reversible illnesses. All 25 participants achieved the ability to sit without support, 23/25 (92%) achieved walking with assistance, and 22/25 (88%) achieved walking independently. Eight infants had adverse events considered possibly related to nusinersen by the study investigators 31,.

The purpose of the study limitations tables (see Tables 7 and 8) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence following each table. Notable study limitations include a relatively short follow-up, which is inadequate to assess the durability of the treatment effect or safety, especially those that are potentially rare or have delayed onset.

Table 5. Key Characteristics Summary of NURTURE Study
Study Study Type Country Sites Dates Participants Interventions Follow-Up
            Active    
De Vivo et al (2019); NURTURE (NCT02386553) 31, Single-arm cohort U.S., EU, Asia 15 2015-ongoing
Presymptomatic infants (N=25) documented to have 5q spinal muscular atrophy homozygous gene deletion, homozygous variant or compound heterozygote variant, and deemed likely to develop SMA type I (n=15) or II (n=10)
Nusinersen at FDA-approved dose   Analysis March 2019: median age 34.8 (25.7 to 45.4], and median time on treatment 2.9 years.
EU: European Union; FDA: U.S. Food and Drug Administration; SMA: spinal muscular atrophy. NURTURE: A Study of Multiple Doses of Nusinersen (ISIS 396443) Delivered to Infants With Genetically Diagnosed and Presymptomatic Spinal Muscular Atrophy
Table 6. Summary of Results of NURTURE Study
Study All 2 SMN2 copy number 3 SMN2 copy number
De Vivo et al (2019); NURTURE31,
N 25 15 10
Primary Endpoint: Survival and respiratory intervention (invasive or non-invasive for ≥6 h per day continuously for ≥7 days or tracheostomy) Not estimablea as there were too few events - -
Secondary endpoints      
Sitting without support (%) 100% 100% 100%
Walking with assistance (%) 92% 87% 100%
HINE-2 motor milestone, mean (range)
Baseline
Post-treatment

Not reported
Not reported

2.7 (0–5)
23.9 (16–26)

3.2 (0–7)
26.0 (26–26)
CHOP-INTEND scores at last visit, mean (range)
% with maximum CHOP INTEND score of 64
Clinically manifested SMA, % (95% CI)
At age of 13 months
At age of 24 months

Not reported
Not reported
Not reported
Not reported

62.1 (48–64)
67%
67 (39 to 87%)
47 (22 to 73%)

63.4 (58–64)
100%
20 (4 to 56%)
0
CHOP-INTEND: Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders; HINE: Hammersmith Infant Neurologic Examination; NURTURE: A Study of Multiple Doses of Nusinersen (ISIS 396443) Delivered to Infants With Genetically Diagnosed and Presymptomatic Spinal Muscular Atrophya All are alive and none required permanent ventilation. Four (16%) infants (all with 2 SMN2 copies) utilized respiratory intervention for ≥6 h per day continuously for ≥7 days during an acute, reversible illness. At the last study day prior to data cutoff, 2 of these infants no longer utilized respiratory intervention; these infants had previously received respiratory intervention for ≥6 h per day for totals of 20 and 266 days during the course of the study. The other 2 infants continued to receive respiratory intervention for 2 and 10 h per day, respectively, at the last study day prior to data cutoff; these infants received respiratory intervention for ≥6 h per day for totals of 236 and 644 days, respectively, over the course of the study. 
Table 7. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Follow-Upe
De Vivo et al (2019); NURTURE31,        
  1. Not sufficient duration for benefit
  2. Not sufficient duration for harms
NURTURE: A Study of Multiple Doses of Nusinersen (ISIS 396443) Delivered to Infants With Genetically Diagnosed and Presymptomatic Spinal Muscular AtrophyThe study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest.c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively.d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported.e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.
Table 8. Study Design and Conduct Limitations
Study Allocationa Blindingb Selective Reportingc Data Completenessd Powere Statisticalf
De Vivo et al (2019); NURTURE31,
  1. Participants not randomly allocated
  2. Allocation not concealed
  3. Allocation concealment unclear
  1. Outcome assessed by treating physician
       
NURTURE: A Study of Multiple Doses of Nusinersen (ISIS 396443) Delivered to Infants With Genetically Diagnosed and Presymptomatic Spinal Muscular AtrophyThe study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment. Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for selection bias.b Blinding key: 1. Not blinded to treatment assignment; 2. Not blinded outcome assessment; 3. Outcome assessed by treating physician.c Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.d Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials).e Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference.f Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4. Comparative treatment effects not calculated.

Section Summary: Presymptomatic Patients with a Genetic Diagnosis of Spinal Muscular Atrophy and a Minimum of 2 But Less Than 4 Copies of SMN2

The evidence for use of nusinersen for presymptomatic type I (infantile-onset) spinal muscular atrophy consists of a single-arm study. After a median follow-up of 2.9 years of 25 infants who received the first dose of nusinersen at a median age of 22 days and are now median age of 34.8 months, 100% were alive, 100% achieved the ability to sit without support, 92% achieved the ability to walk with assistance, and 88% achieved the ability to walk independently. While none required permanent ventilation or tracheostomy, 4 (16%) participants with 2 SMN2 copies utilized respiratory support for ≥6 h/day for ≥7 consecutive days that was initiated during acute, reversible illnesses. These results demonstrate that early treatment resulted in the achievement of motor milestones among patients who are not likely to attain them without treatment. However, the data is limited for the durability of response, and long-term data documenting safety and efficacy are needed.

Summary of Evidence

For individuals who are presymptomatic with a genetic diagnosis of spinal muscular atrophy and a minimum of 2 but less than 4 copies of SMN2 who receive nusinersen, the evidence includes an open-label single-arm trial. Relevant outcomes are overall survival, change in disease status, morbid events, functional outcomes, quality of life, and treatment-related mortality and morbidity. After a median of 2.9 years of follow-up of 25 infants who received the first dose of nusinersen at a median age of 22 days and are now median age of 34.8 months, 100% were alive, 100% achieved the ability to sit without support, 92% achieved the ability to walk with assistance, and 88% achieved the ability to walk independently. While none required permanent ventilation or tracheostomy, 4 (16%) participants with 2 SMN2 copies utilized respiratory support for ≥6 h/day for ≥7 consecutive days that was initiated during acute, reversible illnesses. These results demonstrate that early treatment resulted in the achievement of motor milestones among patients who are not likely to attain them without treatment. However, the data is limited for the durability of response, and long-term data documenting safety and efficacy are needed. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Population

Reference No. 1

Policy Statement

[X] MedicallyNecessary [ ] Investigational

Population Reference No. 2 

Type I (Infantile-Onset) Spinal Muscular Atrophy

The evidence base for infantile-onset or type I spinal muscular atrophy is summarized in Table 9. The supporting EMBRACE and CS3A studies are not reviewed in detail. EMBRACE was terminated early to roll over participants to an open label extension study after the demonstration of motor function benefit with nusinersen in ENDEAR.

Table 9. Summary of Key Clinical Trial in Infantile-Onset or Type I Spinal Muscular Atrophy Patients
Study (Trial) Trial Name Design Dates Patients (N) Outcome
Finkel et al (2017)32, (NCT02193074) ENDEAR RCT Aug 2014 Symptomatic (82) Efficacy, safety
Ascadi et al (2021)33, (NCT02462759) EMBRACE RCT Aug 2015 Symptomatic (21) Safety, tolerability
Finkel et al (2016)19,(NCT01839656) CS3A Single arm May 2013 Symptomatic (20) Safety, PK
EMBRACE: A Study to Assess the Safety and Tolerability of Nusinersen in Participants With Spinal Muscular Atrophy; ENDEAR: A Study to Assess the Efficacy and Safety of Nusinersen (ISIS 396443) in Infants With Spinal Muscular Atrophy; PK: pharmacokinetics; RCT: randomized controlled trial; a Available as abstract only.

Randomized Studies

The pivotal ENDEAR trial was a multicenter randomized, double-blind trial in which 121 infants with a documented genetic diagnosis of spinal muscular atrophy with symptom onset before 6 months of age were randomized 2:1 to nusinersen (n=80) or to sham injection (n=41).32, Nusinersen was approved on the basis of a planned interim analysis of 82 patients who completed at least 183 days of treatment or died or withdrew. Patients’ demographics at baseline were 44% male, and 87% white, with a median length of treatment of 261 days (range, 6-442 days). The primary endpoint was the proportion of motor milestone responders. See Table 10 for the study summary.

Results are summarized in Table 11.32, The trial met its coprimary endpoints, with nusinersen showing clinically meaningful improvement in motor milestones and probability of surviving or receiving permanent-assisted ventilation compared with sham control. The median time to death or the use of permanent-assisted ventilation was 22.6 weeks in the control group and was not reached in the nusinersen group. Multiple secondary endpoints showed a consistency in treatment effect favoring nusinersen over sham control. While no irreversible harms were observed in the preliminary clinical data analyzed by the FDA before drug approval, the FDA noted that such harms could not be ruled out based on animal toxicity data (potential of neurotoxicity) and class effects of antisense oligonucleotides (coagulation abnormalities, thrombocytopenia, renal toxicity). Given the limited data on the durability of response, long-term data documenting safety and efficacy is needed.

The purpose of the study limitations table (see Table 12) is to display notable limitations identified in a study. This information is synthesized as a summary of the body of evidence following each table. No study design and conduct gaps were identified. Notable study limitations include a relatively short follow-up, which is inadequate to assess the durability of the treatment effect or safety, especially those that are potentially rare or have delayed onset. In addition to the gaps identified in the tables, the 2 treatment groups were not balanced with respect to age at symptom onset, use of ventilatory support, and the presence of symptoms specific to spinal muscular atrophy. These were higher in the nusinersen group than the control group. None of these differences were tested for statistical significance.

Table 10. Summary of Key ENDEAR Trial
Study Study Type Country Sites Dates Participants Interventions Follow-Up
            Active Comparator  
Nusinersen                
Finkel et al (2017); ENDEAR32, DB-RCT U.S., EU, Asia 31 2014-2016 SMA type I with symptom onset before 6 mo (N =121) Nusinersen at FDA- approved dose (n=80) Placebo (n=41) Median length of treatment of 261 days (range, 6-442 days); trial terminated early
DB-RCT; double-blind randomized controlled trial; ENDEAR: A Study to Assess the Efficacy and Safety of Nusinersen (ISIS 396443) in Infants With Spinal Muscular Atrophy; EU: European Union; FDA: U.S. Food and Drug Administration; SMA: spinal muscular atrophy. 
Table 11. Summary of Results of ENDEAR Trial
Study Percent motor milestone response (HINE section 2)a No death or use of permanent-assisted ventilationb ≥4-point improvement in CHOP INTEND scorec No death No use of permanent-assisted ventilationb CMAP responsed
Finkel et al (2017); ENDEAR32,          
N 121 121 121 121 121 121
Nusinersen 37/73 (51) 49/80 (61) 52/73 (71) 67/80 (84) 62/80 (78) 26/73 (36)
Sham 0/37 (0) 13/41 (32) 1/37 (3) 25/41 (61) 28/41 (68) 2/37 (5)
HR (95% CI) - 0.53 (0.32 to 0.89) - 0.37 (0.18 to 0.77) 0.66 (0.32 to 1.37) -
P value <.001 .005 <.001 .004 .13 -
CI: confidence interval; CMAP: compound muscle action potential; HINE: Hammersmith Infant Neurologic Examination; HR: hazard ratio; ENDEAR: A Study to Assess the Efficacy and Safety of Nusinersen (ISIS 396443) in Infants With Spinal Muscular Atrophy;Adapted from Finkel et al (2017).32, Values are percent or n (%) or as otherwise indicated. Final analysis conducted on November 21, 2016 included 121 data from infants who had undergone randomization and the assigned procedure at least once.a Motor milestone response was defined according to scores on the HINE-2, which assesses the development of motor function through the achievement of motor milestones; in this trial, the scores accounted for 7 of the 8 motor milestone categories, excluding voluntary grasp. Infants were considered to have a motor milestone response if they met the following 2 criteria: improvement in at least 1 category (ie, an increase in the score for head control, rolling, sitting, crawling, standing, or walking of ≥1 point, an increase in the score for kicking of ≥2 points, or achievement of the maximal score for kicking) and more categories with improvement than categories with worsening (ie, a decrease in the score for head control, rolling, sitting, crawling, standing, or walking of ≥1 point or a decrease in the score for kicking of ≥2 points).b Permanent-assisted ventilation was defined as tracheostomy or ventilatory support for at least 16 h/d for more than 21 continuous days in the absence of an acute reversible event, as determined by an independent endpoint adjudication committee.cA CHOP INTEND response was defined as an increase of at least 4 points from baseline in CHOP INTEND score at the end-of-trial visit (day 183, 302, or 394).dA CMAP response was defined as an increase in the peroneal CMAP amplitude to at least 1 mV (or maintenance of an amplitude of ≥1 mV) at the end-of-trial visit (day 183, 302, or 394).
Table 12. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Follow-Upe
Finkel et al (2017); ENDEAR32,        
  1. Not sufficient duration for benefit
  2. Not sufficient duration for harms
ENDEAR: A Study to Assess the Efficacy and Safety of Nusinersen (ISIS 396443) in Infants With Spinal Muscular AtrophyThe study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest.c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively.d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported.e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.

Section Summary: Type I (Infantile-Onset) Spinal Muscular Atrophy

The evidence for use of nusinersen for symptomatic type I (infantile-onset) spinal muscular atrophy consists of a double-blind RCT. The phase 3 confirmatory ENDEAR trial (N =121) showed clinically meaningful and statistically significant improvement in motor milestones and event-free survival that exceeded those seen in the control group. However, most patients had a modest response or no response, and only a small proportion of patients (6%) gained the ability to sit without assistance. On average, the mean motor milestone score in nusinersen-treated patients improved by 3 points over 6 months. Given the limited data on the durability of response, long-term data documenting safety and efficacy are needed.

Summary of Evidence

For individuals who have type I (infantile-onset) spinal muscular atrophy who receive nusinersen, the evidence includes a randomized, double-blind, controlled trial. Relevant outcomes are overall survival, change in disease status, morbid events, functional outcomes, quality of life, and treatment-related mortality and morbidity. The phase 3 confirmatory ENDEAR trial (N =121) showed clinically meaningful and statistically significant improvement in motor milestones, event-free survival, and overall survival that exceeded those seen in the control group, with an acceptable safety profile. The proportion of patients, who met the primary endpoint responder definition of achieving motor milestones, was 51% in the nusinersen arm compared with 0% in the sham-controlled arm. Further, the hazard ratio for event-free survival was 0.53 favoring nusinersen over sham-controlled. It is notable, however, that 50% of nusinersen-treated subjects did not achieve the primary endpoint motor milestone response. Only a small proportion of patients (6%) gained the ability to sit without assistance. On average, the mean motor milestone score in nusinersen-treated patients improved by 3 points over 6 months. Given the limited data on the durability of response, long-term safety, and lack of efficacy in a substantial number of patients continued risk-benefit assessment of long-term treatment with nusinersen is necessary. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Population

Reference No. 2

Policy Statement

[X] MedicallyNecessary [ ] Investigational

Population Reference No. 3 

Type II or III Spinal Muscular Atrophy

The evidence base for patients with type II or III spinal muscular atrophy is summarized in Table 13. The evidence base for patients with type II and III spinal muscular atrophy consists of 4 early-phase, open-label studies and a double-blind phase 3 RCT. Of these, data from phase 1 single-arm studies are not reviewed in detail because they were early dose-finding and proof-of-concept studies.

Table 13. Summary of Key Trial Characteristics in Type II and III Spinal Muscular Atrophy Patients
Study (Trial) Trial Name Design Dates N Outcomes
Chiriboga et al (2016)34, (NCT01494701) CS1 1 arm Nov 2011 28 Safety and tolerability
Chiriboga et al (2016)34, (NCT01780246) CS10 1 arm Jan 2013 18 Safety and tolerability
Darras et al (2019)35, (NCT01703988) CS2 1 arm Oct 2012 34 Safety and tolerability
Darras et al (2019)35, (NCT02052791) CS12 1 arm Jan 2014 47 Safety and tolerability
Mercuri et al (2018)36, (NCT02292537) CHERISH RCT July 2014 126 Efficacy and safety
CHERISH: A Study to Assess the Efficacy and Safety of Nusinersen in Participants With Later-onset Spinal Muscular Atrophy (SMA); RCT: randomized controlled trial; SMA: spinal muscular atrophy

Randomized Studies

The pivotal CHERISH trial randomized 126 nonambulatory individuals. See Table 14 for the study summary.The primary endpoint was change in Hammersmith Functional Motor Scale Expanded score compared with baseline. Results are summarized in Table 15. The trial met its primary endpoints with nusinersen showing clinically meaningful improvement in mean Hammersmith Functional Motor Scale Expanded scores compared with sham control. In terms of responder analysis, a higher percentage of children in the nusinersen group (57%) than in the control group (26%; p<.001) had an increase from baseline to month 15 in the Hammersmith Functional Motor Scale Expanded score of at least 3 points, which was considered meaningful. Multiple secondary endpoints summarized in Table 15 showed a consistency in treatment effect favoring nusinersen compared with sham control. The overall incidences of adverse events and moderate and serious adverse events were similar for the nusinersen group and the control group (93% and 100% vs 46% and 55%, respectively). Adverse events with an incidence of 5 or more percentage points higher in the nusinersen group than in the control group were pyrexia, headache, vomiting, back pain, and epistaxis. Given the limited data on the durability of response, long-term data documenting safety and efficacy are needed.

The purpose of the study limitations table (see Table 16) is to display notable limitations identified in a study. This information is synthesized as a summary of the body of evidence following each table. No study design and conduct gaps were identified. Notable limitations include a relatively short follow-up, which is inadequate to assess the durability of the treatment effect or safety, especially those that are potentially rare or have delayed onset. In addition, survival, ventilation, and event-free survival were not evaluated.

Table 14. Summary of Key Characteristics of CHERISH Trial
Study Study Type Country Sites Dates Participants Interventions Follow-Up
            Active Comparator  
Nusinersen                
Mercuri et al (2018); CHERISH36, DB-RCT U.S., EU, Asia 24 2014-2017
  • SMA type II (N =126) with genetic documentation of 5q spinal muscular atrophy (a homozygous deletion, variant, or compound heterozygote in SMN1) with the onset of signs and symptoms at more than 6 months and between ages 2 and 12 years at screening as well as the presence of the following features at screening: the ability to sit independently, no history of the ability to walk independently (defined as the ability to walk ≥15 feet unaided), and a Hammersmith Functional Motor Scale Expanded score between 10 and 54.
  • Children were excluded if they had a severe contracture, evidence of severe scoliosis on radiography, respiratory insufficiency, or a gastric tube placed to provide adequate nutrition.
Nusinersen at FDA- approved dose (n=84) Placebo (n=42) Prespecified interim analysis when all children followed for a minimum of 6 months and 39 or more children had completed 15-month evaluations
CHERISH: A Study to Assess the Efficacy and Safety of Nusinersen (ISIS 396443) in Participants With Later-onset Spinal Muscular Atrophy (SMA); DB-RCT; double-blind randomized controlled trial; EU: European Union; FDA: U.S. Food and Drug Administration; SMA: spinal muscular atrophy.
Table 15. Summary of Results of CHERISH Trial
Study D in HFMSE score from baseline, LSM (95% CI)a Patients with D in HFMSE score ≥3 points Percent (95% CI) Patients who achieved ≥ 1 new WHO motor milestone Percent (95% CI) Change from baseline in no. of WHO motor milestones achieved, LSM (95% CI)a Change from baseline in RULM score, LSM (95% CI)a Patients who achieved ability to stand alone Percent (95% CI) Patients who achieved ability to walk with assistance Percent (95% CI)
Mercuri et al (2018); CHERISH36,
N 126 126 126 126 126 126 126
Nusinersen 3.9 (3.0 to 4.9) 57 (46 to 68) 20 (11 to 31) 0.2 (0.1 to 0.3) 4.2 (3.4 to 5.0) 2 (0 to 8) 2 (0 to 8)
Sham -1.0 (-2.5 to 0.5) 26 (12 to 40) 6 (1 to 20) -0.2 (-0.4 to 0) 0.5 (-0.6 to 1.6) 3 (0 to 15) 0 (0 to 10)
Difference (95% CI) 4.9 (3.1 to 6.7) 30.5 (12.7 to 48.3) 14 (-7 to 34) 0.4 (0.2 to 0.7) 3.7 (2.3 to 5.0) -1 (-22 to 19) 2 (-19 to 22)
Odds ratio (95% CI)b - 6 (2 to 15) - - - - -
P value <.001c <.001 - - - - -
CHERISH: A Study to Assess the Efficacy and Safety of Nusinersen (ISIS 396443) in Participants With Later-onset Spinal Muscular Atrophy (SMA); CI: confidence interval; HFMSE: Hammersmith Functional Motor Scale–Expanded; LSM: least-squares mean; RULM: Revised Upper Limb Module; WHO: World Health Organization.Adapted from Mercuri et al (2018).36,Outcomes assessed at 15 Months. In the final analysis, the multiple imputation method was used for missing data to assess changes from baseline in the HFMSE score, percentage of children with a change in HFMSE score of at least 3 points, and change from baseline in the RULM score. The proportion of missing data for 15-month time-point for HFMSE score were 21% (18/84) and 19% (8/42) in the nusinersen and control group, respectively, and imputed using a multiple imputation method to conduct an intention-to-treat analysis.a LSM change and LSM difference in change between groups were based on analysis of covariance with group assignment as a fixed effect and with adjustment for each child’s age at screening and the value at baseline.b This value is an odds ratio rather than a difference. The odds ratio for nusinersen vs control was based on logistic regression, with group assignment as a fixed effect and with adjustment for each child’s age at screening and the HFMSE score at baseline.c Because the p-value for the primary endpoint was significant in the interim analysis, this endpoint was not formally tested for significance in the final analysis.
Table 16. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Follow-Upe
Mercuri et al (2018); CHERISH36,      
  1. Key health outcomes not addressed;
  1. Not sufficient duration for benefit
  2. Not sufficient duration for harms
CHERISH: A Study to Assess the Efficacy and Safety of Nusinersen (ISIS 396443) in Participants With Later-onset Spinal Muscular Atrophy (SMA); The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest.c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively.d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported.e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.

Section Summary: Type II or III Spinal Muscular Atrophy

The evidence for the use of nusinersen for patients with type II or III spinal muscular atrophy consists of a double-blind RCT. The phase 3 confirmatory CHERISH trial (126 patients) showed clinically meaningful and statistically significant improvement in motor milestones that exceeded those seen in the control group. Multiple secondary endpoints showed a consistency in treatment effect favoring nusinersen over sham control. The treatment effect was greater in younger children and in those who received treatment earlier in their disease course. Given the limited data on the durability of response, long-term data documenting safety and efficacy are needed.

Safety: Nusinersen

As per the prescribing label, thrombocytopenia (including acute, severe thrombocytopenia) and renal toxicity (including potentially fatal glomerulonephritis) have been observed with antisense oligonucleotides.14, In the controlled ENDEAR trial, the most common adverse events that occurred in at least 20% of nusinersen-treated patients and occurred at least 5% more frequently than in sham-controlled patients were a lower respiratory infection, upper respiratory infection, and constipation. Atelectasis (a serious adverse event) was more frequent in nusinersen-treated patients (14%) than in sham control (5%). Adverse events reported verbally were not assessable in the sham-controlled trial because patients were infants. In the open-labeled studies of patients with type II or III spinal muscular atrophy, the most common adverse events were a headache (50%), back pain (41%), and post-lumbar puncture syndrome (41%), which occurred within 5 days of lumbar puncture. Other adverse events in these patients were consistent with reactions observed in the controlled study. Also, 1 case of severe hyponatremia in an infant that required salt supplementation for 14 months and 2 cases of rash were reported. Both patients with rash continued to receive nusinersen and had a spontaneous rash resolution.

Development of anti-nusinersen antibodies was assessed in 126 patients of whom 5 (4%) developed treatment-emergent antidrug antibodies, of which 3 were transient, and 2 were persistent. There are insufficient data to evaluate the effect of antidrug antibodies on clinical response, adverse events, or the pharmacokinetic profile of nusinersen.14,

Summary of Evidence

For individuals who have type II or III spinal muscular atrophy who receive nusinersen, the evidence includes a double-blind, randomized controlled trial. Relevant outcomes are overall survival, change in disease status, morbid events, functional outcomes, quality of life, and treatment-related mortality and morbidity. Efficacy findings from single-arm studies of type II and III spinal muscular atrophy are difficult to interpret because these trials used a wide range of nusinersen doses and lacked control arms. The phase 3 confirmatory CHERISH trial (N =126) showed clinically meaningful and statistically significant improvement in motor milestones (measured using Hammersmith Functional Motor Scale–Expanded scores) that exceeded those seen in the control group (difference of 5.9 points favoring nusinersen over sham control, p<.001). The respective proportion of patients with clinically meaningful improvements in Hammersmith scores greater than 3 points was 57% vs 26% (p<.001). Multiple secondary endpoints also showed a consistency in treatment effect favoring nusinersen over sham control. Given the limited data on the durability of response, long-term safety, and lack of efficacy in a substantial number of patients continued risk-benefit assessment of long-term treatment with nusinersen is necessary. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Population

Reference No. 3

Policy Statement

[X] MedicallyNecessary [ ] Investigational

Population Reference No. 4 

Onasemnogene Abeparvovec-Xioi

Clinical Context and Therapy Purpose

The purpose of onasemnogene abeparvovec-xioi in patients who have a genetic diagnosis of spinal muscular atrophy is to provide a treatment option that is an improvement on existing therapies. Potential benefits of this gene therapy37, may include the following:

The following PICO was used to select literature to inform this review.

Populations

The relevant population of interest is individuals who are symptomatic infants diagnosed with type 1 spinal muscular atrophy and presymptomatic infants with a genetic diagnosis of spinal muscular atrophy.

Interventions

The therapy being considered is onasemnogene abeparvovec-xioi.

Comparators

The relevant comparators are continued medical management (respiratory, digestive, and orthopedic support) and nusinersen.

Outcomes

The general outcomes of interest are survival, functional ability, quality of life, and treatment-related mortality and morbidity. For details, see Table 4. Given the heterogeneity and varying life expectancies among patients with different spinal muscular atrophy subtypes, the timing of follow-up of studies to reasonably assess whether onasemnogene abeparvovec-xioi offers a net health benefit will differ by spinal muscular atrophy subtypes as well as by the timing of treatment initiation relative to symptom onset. For details, see Tables 3 and 4.

Study Selection Criteria

Methodologically credible studies were selected using the following principles:

Non Randomized Studies

The clinical development program for onasemnogene abeparvovec-xioi is summarized in Table 17.

Table 17. Summary of the Clinical Development Program for Onasemnogene Abeparvovec-Xioi
Study Phase N Status
Infants under 6 wk (presymptomatic with a genetic diagnosis of SMA and less than 4 copies of SMN2 )
SPR1NT (NCT03505099) 3 44 Completed
Children <6 mo of age (SMA type I)
Pivotal (NCT02122952)38, 1 15 Completed
STR1VE-US trial (NCT03306277)39, 3 21 Completed
STR1VE-EU trial (NCT03461289) 3 30 Completed
START (NCT03421977)a40, 4 15 Ongoing; estimated completion: Dec 2033
Children up to 60 mo of age (SMA type II)
STRONG (NCT03381729) 1 27 Suspended Apr 2024
Patients between 6 mo and 18 y who are ineligible for the other studies
REACH (yet to register)      
PIVOTAL: Gene Transfer Clinical Trial for Spinal Muscular Atrophy Type 1; SPR1NT: Pre-Symptomatic Study of Intravenous Onasemnogene Abeparvovec-xioi in Spinal Muscular Atrophy (SMA) for Patients With Multiple Copies of SMN2; START: Long-Term Follow-up Study for Patients From AVXS-101-CL-101; STRIVE EU: Single-Dose Gene Replacement Therapy Clinical Trial for Patients With Spinal Muscular Atrophy Type 1; STRIVE US: Gene Replacement Therapy Clinical Trial for Patients With Spinal Muscular Atrophy Type 1; STRONG: Study of Intrathecal Administration of Onasemnogene Abeparvovec-xioi for Spinal Muscular Atrophy.a Long-term, safety follow-up study of patients enrolled in NCT02122952.

Symptomatic Spinal Muscular Atrophy TYPE I (Infantile-Onset)

The clinical development program of onasemnogene abeparvovec-xioi for patients with symptomatic spinal muscular atrophy type I includes 4 prospective cohort studies and is summarized in Table 17.; ). These trials enrolled a total of 65 patients with symptomatic spinal muscular atrophy type I. The study characteristics and results are summarized in Tables 18 and 19.

The FDA approval was based on a pooled analysis of 21 patients with 2 copies of SMN2 from the pivotal phase I and STRIVE-US trial with a data analysis cut off of March 2019. Efficacy was established on the basis of survival, and achievement of developmental motor milestones such as sitting without support. Comparison of the results of the ongoing clinical trial to available natural history data of patients with infantile-onset spinal muscular atrophy was the primary evidence for the effectiveness of onasemnogene abeparvovec-xioi. The FDA analysis is summarized in Table 19. The inclusion and exclusion criteria of the phase 3 confirmatory study (STRIVE-US) were the same as the phase 1 dose-finding study.39, The co-primary efficacy outcomes are functional independent sitting for 30 or more seconds at 18 months of age and event-free survival at 14 months of age (defined as the avoidance of either death or need for tracheostomy or ventilation ≥16 hours/day for ≥2 consecutive weeks). Secondary efficacy outcomes are the ability to thrive at 18 months of age, including not receiving nutrition through mechanical support or other nonoral methods, ability to tolerate thin liquids (formal swallowing test), and maintaining weight (>3rd percentile for age and sex) and ability to remain independent of ventilatory support at 18 months of age. Results are summarized in Tables 18 and 19.

Results of an ongoing study to assess long-term safety and durability of response in infants with SMA type 1 with a median time since dosing of 5.2 years (range 4.6 to 6.2 years) have been published40, and summarized in Tables 18 and 19. Seven of 13 were receiving concomitant nusinersen (all 3 patients in the low-dose cohort and 4 of the 10 patients in the therapeutic-dose cohort). The primary objective is to assess safety, and the secondary objective is to determine whether developmental milestones achieved in the phase 1 clinical trial were maintained and new milestones gained. Thirteen of 15 original patients were included in the analysis; 2 patients’ families declined follow-up participation. At the data cutoff on June 11, 2020, the median age of 13 patients followed was 38.9 months. Serious adverse events occurred in 8 patients (62%), none of which resulted in study discontinuation or death. All 10 patients in the therapeutic-dose cohort remained alive and without the need for permanent ventilation. All 10 patients treated with the therapeutic dose maintained previously acquired motor milestones. Two patients attained the new milestone of “standing with assistance” without the use of nusinersen.

While the current evidence for symptomatic type I spinal muscular atrophy patients is limited to patients with 2 copies of SMN2, approximately 20% of type I spinal muscular atrophy patients may have 3 copies of SMN2.5, Given the treatment effect observed in symptomatic patients, it is possible that patients with 3 copies of SMN2 may experience a clinically meaningful benefit. However, there is no published evidence to support such a hypothesis. Further, there is no published data that supports clinical benefit in Type I spinal muscular atrophy patients who are administered onasemnogene abeparvovec-xioi after 6 months of age.

Novartis Gene Therapies has sponsored creation of RESTORE, a prospective, multicenter, multinational observational registry. 41, Participants will be managed according to usual clinical practice. Both newly recruited SMA treatment centers and sites involved in existing SMA registries will be eligible to participate; de novo; sites already participating in another registry may be included via consortium agreements. Participants will be enrolled over a 5-year period and followed for 15 years or until death. Assessments will include SMA history and treatment, pulmonary, nutritional, and motor milestones, healthcare resource utilization, work productivity, activity impairment, adverse events, quality of life, caregiver burden, and survival. Results for 117 participants describing real-world outcomes in individuals with SMA aged ≥6 months at the time of onasemnogene abeparvovec infusion have been presented at conferences but have not been published in a peer reviewed journal at the time of 2023 update of this review.

Table 18. Summary of Key Characteristics of Nonrandomized Trials
Study Study Type Country Dates Participants Treatment Follow-Up
Mendell et al (2017)38, Single Cohort U.S. 2014-2017 Infants <9 monthsa with biallelic SMN1 deletions or variants with 2 copies of SMN2 (n=15). Patients with c.859G>C variant in SMN2 exon 7 were excluded from the study.b Onasemnogene abeparvovec-xioi (3 minimally effective dosec; 12 proposed therapeutic dosed) Median, 30.7c and 27.8d monthse
Day et al (2021)39, Single Cohort U.S. 2017-2019 Infants <6 months with biallelic SMN1 deletions or variants with 1 or 2 copies of SMN2 (N =22). Patients with c.859G>C variant in SMN2 exon 7 were included in the study.b Onasemnogene abeparvovec (single IV dose of 1.1 X 1014 vector genomes per kg) 18 months
Mendell et al (2021)40, Single Cohort U.S. 2017-2021 Infants <9 monthsa with biallelic SMN1 deletions or variants with 2 copies of SMN2 (n=13). Patients with c.859G>C variant in SMN2 exon 7 were excluded from the study.b No treatment; intent was long-term follow-up for safety Median time since dosing 5.2 years (range, 4.6 to 6.2)
a Protocol was amended to lower the age to 6 months of age or younger.b c.859G>C substitution is a positive modifier and has been shown to results in a mild SMA phenotype.42,c At 6.7×1013 vg/kg. d At 2.0×1014 vg/kg.e The oldest patients is 46.2 months of age, with 40.6 months of follow-up.
Table 19. Summary of Key Results of Nonrandomized Trials
Study Survival Change in Mean CHOP INTEND Score Patients With CHOP INTEND Score >40, n (%) Others Safety
FDA Label (as of the March 2019 data cutoff)43,          
N 21       44
Onasemnogene abeparvovec-xioi 90.5% NR NR
  • 1 patient died at age 7.8 months due to disease progression
  • 1 patient withdrew from the study at age 11.9 months
  • 68% (13 of the 19) patients continuing in the trial reached 14 months of age without permanent ventilation
  • 47.6% (10 of 21) sit without support for ≥ 30 seconds between 9.2 and 16.9 months of age (mean age 12.1 months)
  • 84% (16 of 19) did not require daily non-invasive ventilator use
Elevated ALT/AST a (> ULN): 12 (27.3%)
Vomiting: 3 (6.8%)
Day et al (2021)39,          
N 22        
Onasemnogene abeparvovec-xioi 95% NR 21 (95%)
  • 59% (13 of 22 patients achieved functional independent sitting for 30 s or longer at the 18 months of age study visit)
  • 41% (9 of 22 patients maintained the ability to thriveb at the 18 months of age study visit)
  • All patients had at least 1 adverse event (most common was pyrexia).
    Frequently reported serious adverse events were bronchiolitis, pneumonia,
  • respiratory distress, and respiratory syncytial virus bronchiolitis.
  • 3 serious adverse events were related or possibly related to the treatment (2 cases of elevated hepatic aminotransferases and 1 of hydrocephalus).
Historical cohort (PNCR)9, 20% NR Rare
  • None of the 23 untreated patients achieved functional independent sitting for 30 s or longer at the 18 months of age in the PNCR cohort
  • Ability to thrive data was not reported for the PNCR cohort
  • NA
Mendell et al (2021)40,          
N 13        
Onasemnogene abeparvovec-xioi 100% (10 of 10 in therapeutic dose)
100% (3 of 3 in low dose)
NA NA
  • 7 of 13 receiving concomitant nusinersen (all 3 in the low-dose cohort and 4 of the 10 patients in the therapeutic-dose cohort)
  • None of the 10 in the therapeutic-dose cohort require permanent ventilation
  • 2 of 3 in the low dose remain free of permanent ventilation
  • All 10 patients in therapeutic dose
    cohort maintained previously acquired motor milestones. Two attained the new milestone
    of “standing with assistance” without the use of nusinersen.

Serious adverse events (n=8; 62%)

  • Acute respiratory failure (4 [31%])
  • Pneumonia (4 [31%])
  • Dehydration (3 [23%])
  • Respiratory distress (2 [15%])
  • Bronchiolitis (2 [15%])
AE: adverse events; ALT: alanine aminotransferase; AST; aspartate aminotransferase; CHOP INTEND: Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders; NA: not applicable; NR: not reported; SAE: serious adverse event; ULN: upper limit of normal.a  In the completed clinical trial, 1 patient (the first patient infused in that study) was enrolled prior to the protocol amendment instituting administration of prednisolone before and after infusion.b  Ability to thrive was a composite endpoint defined by swallowing function (the ability to tolerate thin liquids shown by a formal clinical swallowing assessment [eg, bedside swallow exam]) AND nutritional support (feeding exclusively by mouth, defined as not receiving nutrition through a feeding tube or other non-oral methods) AND and weight maintenance (maintaining weight greater than the third percentile for the appropriate age and sex)

The purpose of the study limitations tables (see Tables 20 and 21) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence following each table. Due to strict inclusion criteria, the patient population included in the trial was more homogeneous (eg, SMN2 copy number differences), younger, and treated earlier from the time of onset of symptoms than patients in routine clinical practice. For example, the weighted mean (standard deviation) age of symptom onset and age of confirmed genetic diagnosis in spinal muscular atrophy type I patients was 2.5 (0.6) and 6.3 (2.2) months, respectively, based on a systematic literature review of studies published between 2000 and 2014.44, The weighted diagnostic delay in this systematic review was 3.6 months. In the onasemnogene abeparvovec-xioi phase I study, the age of symptom onset and age of confirmed genetic diagnosis were 1.4 months (range, 0-3 months) and 2 months (range, 0-4.5 months), respectively. Therefore, the benefits observed in this study setting might not translate to patients in a real-world setting. However, with increasing efforts toward newborn screening for spinal muscular atrophy, it is possible that the delay in diagnosis of spinal muscular atrophy may be shortened.

Table 20. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Follow-Upe
Mendell et al (2017)38,  

 

 

 

1. Not sufficient duration for benefit (long-term)
2. Not sufficient duration for harms (long-term)
Day et al (2021)39,  

 

 

 

1. Not sufficient duration for benefit (long-term)
2. Not sufficient duration for harms (long-term)
Mendell et al (2021)40,   4. Not the intervention of interest (7 out of 13 patients received continue to receive nusinersen after receiving one-time gene therapy).

 

 

 
The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest.c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively.d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported.e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.
Table 21. Study Design and Conduct Limitations
Study Allocationa Blindingb Selective Reportingd Data Completenesse Powerd Statisticalf
Mendell et al (2017)38, 1. Participants not randomly allocated
 
3. Outcome assessed by treating physician        
Day et al (2021)39, 1. Participants not randomly allocated 3. Outcome assessed by treating physician        
Mendell et al (2021)40, 1. Participants not randomly allocated 3. Outcome assessed by treating physician   5. Two of the 13 in the high dose cohort did not participate    
The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for selection bias.b Blinding key: 1. Not blinded to treatment assignment; 2. Not blinded outcome assessment; 3. Outcome assessed by treating physician.c Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.d Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials).e Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference.f Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4.Comparative treatment effects not calculated.

Section Summary: Symptomatic Spinal Muscular Atrophy Type I (Infantile-Onset)

The evidence for use of onasemnogene abeparvovec-xioi for symptomatic spinal muscular atrophy type I (infantile-onset) consists of 2 single-arm studies. The FDA approval was based on a pooled analysis of 21 patients from the 2 single-arm studies. The observed treatment effect on survival, event-free survival and achievement of motor functions is beyond what is typical based on the known natural history of patients with spinal muscular atrophy type I with 2 copies of SMN2. Results of the phase 3 confirmatory study (STRIVE-US) published after the FDA approval were largely consistent with previously available findings at the time of approval. Results of an ongoing study to assess long-term safety and durability of response in infants with SMA type 1 with a median time since dosing of 5.2 years showed that the developmental milestones achieved in the phase 1 clinical trial were maintained and new milestones gained. Thirteen of 15 original patients were included in the analysis. All 10 patients in the therapeutic-dose cohort remained alive and without the need for permanent ventilation. All 10 patients treated with the therapeutic dose maintained previously acquired motor milestones. Two patients attained the new milestone of “standing with assistance” without the use of nusinersen. However, 7 of the 13 subsequently received concomitant nusinersen.

Summary of Evidence

For individuals who have spinal muscular atrophy type I (infantile-onset) who receive onasemnogene abeparvovec-xioi, the evidence includes 2 single-arm studies. Relevant outcomes are overall survival, change in disease status, functional outcomes, quality of life, and treatment-related mortality and morbidity. The FDA approval was based on a pooled analysis of 21 patients from the 2 single-arm studies. The observed treatment effect on survival, event-free survival, and achievement of motor functions is beyond what is typical based on the known natural history of patients with spinal muscular atrophy type I with 2 copies of SMN2. Results of the phase 3 confirmatory study (STRIVE-US) published after the FDA approval were largely consistent with previously available findings at the time of approval. Results of an ongoing study to assess long-term safety and durability of response in infants with SMA type 1 with a median time since dosing of 5.2 years showed that the developmental milestones achieved in the phase 1 clinical trial were maintained and new milestones gained. Thirteen of 15 original patients were included in the analysis. All 10 patients in the therapeutic-dose cohort remained alive and without the need for permanent ventilation. All 10 patients treated with the therapeutic dose maintained previously acquired motor milestones. Two patients attained the new milestone of “standing with assistance” without the use of nusinersen. However, 7 of the 13 subsequently received concomitant nusinersen. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Population

Reference No. 4

Policy Statement

[X] MedicallyNecessary [ ] Investigational

Population Reference No. 5 

Presymptomatic Infants with a Genetic Diagnois of Spinal Muscular Atrophy and Less Than 4 Copies of SMN2

The clinical development program of onasemnogene abeparvovec-xioi for presymptomatic infants with a genetic diagnosis of spinal muscular atrophy and less than 4 copies of SMN2 includes a single prospective cohort study called SPR1NT. The study characteristics are summarized in Table 22 and results are summarized in Table 23.The study included 2 cohorts: 14 children with 2 copies of SMN2 and 14 children with 3 copies of SMN2 expected to develop SMA type 1. Results of the cohorts were reported in 2 separate publications.45,46, All patients with 2 copies of SMN2 reached 18-months-of-age (study end) and achieved the primary endpoint of sitting without support for at least 30 seconds.45, Seventy-nine percent (11/14) achieved this milestone within an age-appropriate time period based on the WHO MGRS of healthy children living under conditions highly unlikely to constrain growth.47,All patients with 3 copies of SMN2 reached 24-months-of-age (study end) and achieved the primary endpoint of standing without support up to the 24-months-of-age visit. Ninety-three percent (14/15) achieved this milestone within an age-appropriate time period.46,In the natural history of SMA Type 1, untreated children with 2 copies of the SMN2 backup gene would not achieve such skills. Among the cohort of infants with 3 copies of SMN2 gene, 27% (4/15) stood for 3 seconds or more without assistance, 60% (9/15) stood for 10 seconds or more with assistance and 13% (2/15) walked 5 steps or more without assistance as measured by WHO-MGRS. All patients (29/29) did not need temporary breathing support and remained free of feeding support and 97% (28/29) remained within a normal weight range (3rd-97th percentile for age).

The purpose of the study limitations tables (see Tables 24 and 25) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence following each table. Notable study limitations include a relatively short follow-up, which is inadequate to assess the durability of the treatment effect or safety, especially those that are potentially rare or have delayed onset.

Table 22. Summary of Key Nonrandomized Trials
Study Study Type Country Dates Participants Treatment Follow-Up
Strauss et (202245,) and Strauss et all (2022; SPR1NT)46, Single arm U.S., Europe, Asia 2018- 2021
  • Presymptomatic infants with biallelic deletions of SMN1 and 2 (n=14) or 3 copies (n=15) of SMN2 (N = 29)
  • The primary endpoint among infants with 2 copies of SMN2 is the proportion of patients with functional independent sitting for 30 or more seconds by 18 months of age and among those with 3 copies of SMN2 is the proportion of patients able to stand without support for 3 or more seconds up to 24 months of age.
Single dose intravenous infusion of onasemnogene abeparvovec-xioi at 1.1 X 1014 vector genomes/kg 18 or 24 of age, dependent upon respective SMN2 copy number
 

Table 23. Summary of Key Results of Nonrandomized Trial

Study Survival Change in Mean (±SD) CHOP INTEND Score Patients With CHOP INTEND>40, n(%) Others Safety
Strauss et al (2022)45,          
N 14        
Onasemnogene abeparvovec-xioi 100% 1 month: 3.9 (±8.3)
3 months: 11.2 (±8.8)
6 months: 14.8 (±8.1)
14 (100%)
  • All 14 (100%) children achieved independent sitting for at least 30 seconds at any visit up to 18 months of age
  • All 14 (100%) children achieved motor milestones as defined by BSID and WHO-MGRS
  • 11 of 14 (79%) children stood alone without assistance based on BSID definition and 9 children (64%) walked independently
  • No child required mechanical respiratory support during the trial
  • No serious adverse events were considered treatment-related
    by the investigator
  • 1 patient had serum aminotransferase enzyme concentration >3x ULN. Resolved with prednisolone dose modification
Strauss et al (2022) 46,          
N 15        

Onasemnogene abeparvovec-xioi

100%

NA

NA

  • All 15 (100%) children stood independently based on BSID definition by 24 months of age and all maintained this motor milestone at the 24-month study visit
  • 14 (93%) of 15 children sat independently based on BSID definition by 24 months of age
  • 14 (93%) of 15 children walked independently for at least 5 steps based on BSID definition by 24 months of age
    • The walk of one child was observed during the 24-month study visit but was not recorded. Per study protocol, the child was judged not to have achieved the motor milestone due to absence of recording.
  • No child required mechanical respiratory support during the trial
  • No serious adverse events were considered treatment-related
    by the investigator
BSID: Bayley-III Scales of Infant and Toddler Development; CHOP INTEND: Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders; ITT: Intention-to-treat; NA: not applicable; NR: not reported; SAE: serious adverse event; SD: standard deviation; TULN: upper limit of normal; WHO-MGRS: World Health Organization Multicentre Growth Reference Study.

Table 24. Study Relevance Limitations

Study Populationa Interventionb Comparatorc Outcomesd Follow-Upe
Strauss et (202245,) and Strauss et all (2022; SPR1NT)46,         1. Not sufficient duration for benefit (long-term)
2. Not sufficient duration for harms (long-term)
The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest.c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively.d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported.e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.

Table 25. Study Design and Conduct Limitations

Study Allocationa Blindingb Selective Reportingd Data Completenesse Powerd Statisticalf
Strauss et (202245,) and Strauss et all (2022; SPRINT)46, 1. Participants not randomly allocated
 
3. Outcome assessed by treating physician        
The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for selection bias.b Blinding key: 1. Not blinded to treatment assignment; 2. Not blinded outcome assessment; 3. Outcome assessed by treating physician.c Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.d Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials).e Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference.f Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4.Comparative treatment effects not calculated.

Section Summary: Presymptomatic Infants with a Genetic Diagnosis of Spinal Muscular Atrophy and Less Than 4 Copies of SMN2

The evidence for use of onasemnogene abeparvovec-xioi for presymptomatic infants with a genetic diagnosis of SMA and less than 4 copies of SMN2 consists of a single-arm study (SPR1NT). This trial included infants less than 6 weeks of age who did not display any symptoms of SMA at the time of infusion. The trial was divided into 2 cohorts based on SMN2 copy number. All patients irrespective of SMN2 copy numbers achieved their respective primary endpoint, able to sit without support for ≥30 seconds at 18 months of age for 2 copies of SMN2 or ability to stand for 3 seconds or more without assistance at 24 months for those with 3 copies of SMN2. Multiple secondary endpoints were also supportive of clinical benefit including achievement of age-appropriate gross motor milestones and functions and independence from nutritional and respiratory support. In the natural history of SMA type 1, untreated children with 2 or 3 copies of the SMN2 backup gene would not achieve such skills. These results demonstrate that early treatment resulted in the achievement of motor milestones among patients who are not likely to attain them without treatment.

Summary of Evidence

For individuals who are presymptomatic with a genetic diagnosis of spinal muscular atrophy and less than 4 copies of SMN2 who receive onasemnogene abeparvovec-xioi, the evidence includes one single-arm study- SPR1NT. Relevant outcomes are overall survival, change in disease status, functional outcomes, quality of life, and treatment-related mortality and morbidity. The SPR1NT trial included infants less than 6 weeks of age who did not display any symptoms of SMA at the time of infusion. The trial was divided into 2 cohorts based on SMN2 copy number. All patients irrespective of SMN2 copy numbers achieved their respective primary endpoint, able to sit without support for at least 30 seconds at 18 months of age for 2 copies of SMN2 or ability to stand for 3 seconds or more without assistance at 24 months for those with 3 copies of SMN2. Multiple secondary endpoints were also supportive of clinical benefit including achievement of age-appropriate gross motor milestones and functions and independence from nutritional and respiratory support. In the natural history of SMA Type 1, untreated children with 2 or 3 copies of the SMN2 backup gene would not achieve such skills. These results demonstrate that early treatment resulted in the achievement of motor milestones among patients who are not likely to attain them without treatment. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Population

Reference No. 5

Policy Statement

[X] MedicallyNecessary [ ] Investigational

Population Reference No. 6 

Risdiplam

Clinical Context and Therapy Purpose

The purpose of risdiplam in patients who have spinal muscular atrophy type is to provide a treatment option that is an improvement on existing therapies. Potential benefits of this therapy may include the following:

The following PICO was used to select literature to inform this review.

Populations

The relevant population of interest is individuals with a genetic diagnosis of SMA. Because risdiplam is being studied in clinical trials with participants ranging from birth to 60 years of age, in both infantile-onset and later-onset SMA, a large number of patients with SMA may be potential candidates for treatment.‎

Interventions

The therapy being considered is risdiplam.

Comparators

The relevant comparators are continued medical management (respiratory, digestive, and orthopedic support), nusinersen, and onasemnogene abeparvovec-xioi.

Outcomes

The general outcomes of interest are survival, functional ability, quality of life, and treatment-related mortality and morbidity. Given the heterogeneity and varying life expectancies among patients with different spinal muscular atrophy subtypes, the timing of follow-up of studies to reasonably assess whether risdiplam offers a net health benefit will differ by spinal muscular atrophy subtypes as well as by the timing of treatment initiation relative to symptom onset. For details, see Tables 3 and 4.

Study Selection Criteria

Methodologically credible studies were selected using the following principles:

Non Randomized Studies

The clinical development program for risdiplam is summarized in Table 26.

Table 26. Summary of the Clinical Development Program for Risdiplam in Patients with SMA
Trial NCT Phase Description N Design Status
Infantile-Onset SMA (Type 1); Age at enrollment: 1 to 7 months
FIREFISH NCT02913482 2/3 Part 1: Dose finding and safety
Part 2: Efficacy and Safety
Part 1 (21)
Part 2 (41)
Open-label single arm Ongoing and published48,49,50,
Later-Onset SMA (Type 2 and 3); Age at enrollment: 2 to 25 years
SUNFISH NCT02908685 2/3 Part 1: Dose finding and safety
Part 2: Efficacy and Safety
Part 1 (51)
Part 2 (180)
Randomized, double-blind, placebo-controlled Ongoing and published51,
Treatment-Experienced Patients with SMA; Age at enrollment: 6 months to 60 years
JEWELFISH NCT03032172 2 Safety and tolerability 174a Open-label single arm Ongoing and unpublished (expected completion Dec 2024)
Pre-symptomatic SMA; Birth to 6 weeks
RAINBOWFISH NCT03779334 2 Efficacy and safety 25b Open-label single arm Ongoing and unpublished (expected completion Jun, 2023)
NCT: national clinical trial.SMA: spinal muscular atrophya Enrollment completed with final enrollment of 174 patients (source: clinicaltrials.gov)b Target enrollment

Spinal Muscular Atrophy Type I (Infantile-Onset)

The evidence for type I spinal muscular atrophy (SMA) patients includes an open-label single-arm FIREFISH study (dose-finding phase n=21 and confirmed-dose n=41) in which infants aged between 1 to 7 months with 2 copies of SMN2 gene were treated with risdiplam. After an initial dose-finding protocol (part-1) under which multiple ascending doses of risdiplam were administered (low dose=4; recommended FDA dose=17), patients in part-2 of the study were administered the selected dose and will be followed for 24 months. Patient characteristics and results are summarized in Tables 27 and 28. The primary outcome for part 2 was defined as the percentage of infants who could sit without support at 12-months assessed by Gross Motor Scale of the Bayley Scales of Infant and Toddler Development (Third Edition) (BSID-III).

Results at 12-months and 24-months follow-up have been reported. At 12 months, 29% (12 of 41) were able to sit without support for at least 5 seconds as assessed by BSID-III and at 24 months, 61% (25 of 41) met this primary endpoint. At 12 and 24 months of treatment with risdiplam, 85% (35/41) and 83% (34/41) of trial participants were alive without permanent ventilation.48,49, These results indicate a clinically meaningful deviation from the natural history of untreated infantile-onset SMA. As described in the natural history of untreated infantile-onset SMA, patients would not be expected to attain the ability to sit independently, and no more than 25% of these patients would be expected to survive without permanent ventilation beyond 14 months of age.19,38,

The purpose of the limitations tables (Tables 29 and 30) is to display notable limitations identified in each study. Notable limitations include a relatively short follow-up of 1 year and a small sample size, which is inadequate to precisely assess the magnitude of treatment effect, its durability and safety (especially adverse events that are potentially rare or have delayed onset). To address the lack of evidence on long-term efficacy and safety (beyond 1 year), registries or long-term follow-up should universally enroll all patients that receive risdiplam, as optional enrollment in longer follow-up studies is prone to selection bias. In absence of direct head-to-head comparisons with other FDA-approved treatment for type I SMA such as potentially curative 1 time onasemnogene abeparvovec or lifelong chronic intrathecal nusinersen, the role of risdiplam in the treatment pathway of newly diagnosed treatment naïve type I SMA patients is unclear. It is unknown if risdiplam should be used as a first-line agent or second-line agent in such patients or as a combination treatment. Further, it is unclear, under what clinical circumstances use of risdiplam as a second-line or third-line agent will be justified.

Table 27. Summary of Pivotal Trial Characteristics of Risdiplam for Infantile-Onset SMA
Study Study Type Country Dates Participants Treatment Follow-Up
FIREFISH- Part 1 50, Open-label, single-arm US, EU, South America, Middle East (30 sites) 2016-ongoing
  • Type 1 SMA
  • Age 1-7 months
  • 2 SMN2 copies
  • Confirmed genetic diagnosis of 5q-autosomal recessive SMA
  • Primary endpoints: 1) ability to sit without support for at least 5 secondsa, 2) survival without permanent ventilationb

Patient characteristics

  • Female: 15 (71%)
  • Median age at onset of symptoms: 2.0 mo (0.9-3)
  • Median age at enrollment: 6.7 mo (3.3-6.9)
  • Median time between onset of symptoms and first dose: 4.0 mo (2.0-5.8)
  • Part 1 (2 dose cohort; high dose=17 and low dose=4)
Part 1: Median duration of treatment 14.8 months (range:0.6 to 26.0) and 19 patients treated for a minimum duration of 12 months
FIREFISH- Part 248, Open-label, single-arm with historical control US, EU, South America, Middle East 2016-ongoing

Inclusion and exclusion same as above

Patient characteristics

  • Female: 22 (54%)
  • Median age at onset of symptoms: 1.5 mo (1-3)
  • Median age at enrollment: 5.3 mo (2.2-6.9)
  • Part 2 (risdiplam daily, administered orally 0.2 mg/kg for infants older than 5 months of age and 0.04 or 0.08 mg/kg and adjusted to 0.2 mg/kg within 1 to 3 months after the start of treatment)
24 months
EU: Europe; SMA: spinal muscular atrophy.a Measured by Item 22 of the Bayley Scales of Infant and Toddler Development – Third Edition.b Permanent ventilation defined as requiring a tracheostomy or more than 21 consecutive days of either non-invasive ventilation (≥ 16 hours per day) or intubation, in the absence of an acute reversible event.
Table 28. Summary of Results of Pivotal Trial of Risdiplam for Infantile-Onset SMA
Study Motor Functions Other Survival
FIREFISH a  
Part 1a15,50, N=21 N=21 N=21
Risdiplam
  • BSID-III (Item 22): 7/19 (41%) of all subjects and 7/17 (41%) of cohort B were able to sit without support for at least 5 seconds after 12 months of treatment
  • CHOP-INTEND score of 40 or higher: 11/21 (52%)
At month 12, 86% (3 in the low-dose and 15 in the high-dose cohort) were able to feed orally, either exclusively or in combination with a feeding tube.
  • After 12 month of treatment: 19/21 (90.5%)b,c
  • After 23 month of treatment: 17/21 (81%)b,d
Part 2 (1-year results)e48, N=41   N=41
Risdiplam
  • BSID-III (Item 22): 12 (29%) able to sit without support for at least 5 seconds after 12 months of treatment
  • CHOP-INTEND score of 40 or higher: 23 (56%)
  • Increase of ≥4 points from baseline in the CHOP-INTEND score: 37 (90%)
  • HINE-2 motor-milestone response: 32 (78%)

NA

35 (85%)b after 12 months of treatment
Historical Control
  • BSID-III (Item 22): Typically untreated infants are unable to sit without support for at least 5 seconds
  • CHOP-INTEND score of 40 or higher: 17%
  • Increase of ≥4 points from baseline in the CHOP-INTEND score: 17%
  • HINE-2 motor-milestone response: 12%

NA

42%
Part 2 (2-year results)49, N=41   N=41
Risdiplam
  • BSID-III (Item 22): 25 (61%) able to sit without support for at least 5 seconds after 24 months of treatment
  • CHOP-INTEND score of 40 or higher: 31 (76%)
  • Increase of ≥4 points from baseline in the CHOP-INTEND score: 37 (90%)
  • HINE-2 motor-milestone response: 35 (85%)

NA

34 (83%)b after 24 months of treatment
BSID: Bayley Scales of Infant and Toddler Development (Third Edition); CHOP INTEND: Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders; HINE: Hammersmith Infant Neurologic Examination; NA: not available; SMA: spinal muscular atrophy. a In Part 1 of the study, 21 patients have been treated with risdiplam for over 12 months (4 patients received low dose [Cohort A] and 17 received FDA recommended dose of 0.2 mg/kg/day risdiplam [Cohort B].b Defined as alive with no permanent ventilation (i.e. no tracheostomy or BiPAP ≥16 hours per day continuously for >3 weeks or continuous intubation >3 weeks, in the absence of, or following the resolution of an acute reversible event).c Reached 15 months of age or olderd Reached an age of 28 months or older; median 32 months; range 28 to 45 months.e In Part 2 of the study, 41 patients were treated with risdiplam for over 12 months with FDA recommended dose of 0.2 mg/kg/day risdiplam
Table 29. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Follow-Upe
FIREFISH50,48,     1. Not standard or optimal (lack of head to head comparisons with current standard of care)   1. Not sufficient duration for benefit
2. Not sufficient duration for harms
The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest.c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively.d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported.e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.
Table 30. Study Design and Conduct Limitations
Study Allocationa Blindingb Selective Reportingc Data Completenessd Powere Statisticalf
FIREFISH50,48, 1. Participants not randomly allocated
2. Allocation not concealed
3. Allocation concealment unclear
3. Outcome assessed by treating physician        
The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for selection bias.b Blinding key: 1. Not blinded to treatment assignment; 2. Not blinded outcome assessment; 3. Outcome assessed by treating physician.c Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.d Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials).e Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference.f Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4. Comparative treatment effects not calculated.

Section Summary: Spinal Muscular Atrophy Type I (Infantile-Onset)

The evidence for use of risdiplam for symptomatic SMA type I (infantile-onset) individuals consists of a single-phase 2/3 FIREFISH study. Results of part 1 (dose-finding and exploratory) of the study in which 4 patients received low-dose and 17 patients who received FDA recommended dose were the basis for FDA approval. Results showed 41% (7 of 17 infants) were able to sit without support for at least 5 seconds as assessed by BSID-III after 12 months of treatment. After a minimum follow-up of 12 and 23 months of treatment with risdiplam, 90% (19/21) and 81% (17/21) of patients were alive without permanent ventilation. Results of part 2 of the study in which 41 patients received FDA recommended dose reported that after 12 and 24 months of treatment, 29% and 61% were able to sit without support for at least 5 seconds, respectively, 56% and 76% achieved a CHOP-INTEND score of 40 or higher, respectively and 85%, and 83% survived without permanent ventilation, respectively. The observed treatment effect on motor functions and event-free survival is beyond what is typical based on the known natural history of patients with SMA type I with 2 copies of SMN2.

Summary of Evidence

For individuals who have symptomatic SMA type I (infantile-onset) who receive risdiplam, the evidence includes 1 single-arm prospective phase 2/3 FIREFISH study. Relevant outcomes are overall survival, change in disease status, functional outcomes, health status measures, quality of life, and treatment-related mortality and morbidity. Results of a part 1 study (dose-finding and exploratory) in which 4 patients received low dose and 17 patients received high dose risdiplam are available. The results of the part 1 study (dose-finding and exploratory) showed 41% (7 of 17 infants) were able to sit without support for at least 5 seconds as assessed by BSID-III after 12 months of treatment. After a minimum follow-up of 12 and 23 months of treatment with risdiplam, 90% (19/21) and 81% (17/21) of patients were alive without permanent ventilation. Results of a part 2 study in which 41 patients received the FDA recommended dose reported that after 12 months of treatment, 29% were able to sit without support for at least 5 seconds, 56% achieved a CHOP-INTEND score of 40 or higher, 90% achieved an increase of at least 4 points from baseline in the CHOP-INTEND score, 78% achieved a HINE-2 motor milestone response, and 85% survived without permanent ventilation.Results of part 2 of the study in which 41 patients received FDA recommended dose reported that after 12 and 24 months of treatment, 29% and 61% were able to sit without support for at least 5 seconds respectively, 56% and 76% achieved a CHOP-INTEND score of 40 or higher respectively and 85% and 83% survived without permanent ventilation respectively. The observed treatment effect on motor functions and event-free survival is beyond what is typical based on the known natural history of patients with SMA type I with 2 copies of SMN2. However, there is limited data to assess the long-term durability of treatment effect as well as safety-related to adverse events that are rare or have delayed onset. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Population

Reference No. 6

Policy Statement

[X] MedicallyNecessary [ ] Investigational

Population Reference No. 7

Spinal Muscular Atrophy Type II and III

The evidence for pediatric or adult patients (2-25 years old) with type II or III SMA patients includes the ongoing double-blind, 2-part, placebo-controlled SUNFISH study. Patient characteristics and results are summarized in Tables 31 and 32.

Part 1 was dose-finding and exploratory in 51 patients for 12 weeks. Part 2 was the randomized, double-blind, placebo-controlled in which patients received risdiplam or placebo for 12 months. After 12 months, placebo recipients were switched to risdiplam for the next 12 months while those receiving risdiplam continued to receive treatment until 24 months.51,This trial included a patient population who previously were excluded from clinical trials, such as ages up to 25, severe scoliosis, and low baseline function. At baseline, 67% of patients had scoliosis (32% of them with severe scoliosis). Results of Part 2 are discussed next. The least-square mean change in MFM-32 from baseline was greater in patients receiving risdiplam compared with placebo with a difference of 1.55 (95% CI: 0.30 to 2.81). Responder analysis (using a threshold of increase in 3 or more points in MFM-32 as being clinically meaningful) showed that 38% of those receiving risdiplam met the criteria of achieving a clinically meaningful improvement in motor function compared to 24% in the placebo arm. Clinical benefit was also observed in the upper limb function as measured by the revised upper limb module (RULM). Risdiplam treated patients had a mean 1.6 point improvement from baseline at month 12 compared to no mean score change in the placebo group (p=.047). In general, increases on motor function tests would not be expected in the natural history of Type 2 and 3 SMA, and the differences observed between patients treated with risdiplam and those on placebo are considered clinically meaningful.28,

Subgroup analysis of differences in MFM-32 and RULM total scores by age, SMA subtype, and SMN2 copy number are summarized in Table 33. Planned subgroup analysis of MFM-32 and RULM by age showed a trend for greater efficacy in the younger age group. This is consistent with the pathophysiology of SMA in which motor neurons are progressively lost with time.

For MFM-32, subgroup analysis based on SMA subtype showed treatment differences in favor of risdiplam versus placebo in both SMS types 2 and 3. Subgroup analysis based on SMN2 copy number showed treatment differences in favor of risdiplam versus placebo based upon point estimates in individuals with 3 and 4 copies. However, the confidence intervals were wide, and specifically for individuals with SMN2 copy number 4, the confidence interval was inclusive of negative treatment effect. There were only 4 participants with 2 SMN2 2 copy numbers and there was a negative treatment effect. All subgroup analyses were not powered to detect statistically significant differences.

For RULM subgroup analysis was similar. The subgroup analysis based on the SMA subtype also showed treatment differences in favor of risdiplam versus placebo in both types 2 and 3. Subgroup analysis based on SMN2 copy number showed treatment differences in favor of risdiplam versus placebo based upon point estimates in individuals with 3 and 4 copies. However, the confidence intervals were wide, and specifically for individuals with SMN2 copy number 4, the confidence interval was inclusive of negative treatment effect. There were only 4 participants with 2 SMN2 copy numbers and there was a negative treatment effect. All subgroup analyses were not powered to detect statistically significant differences.

The purpose of the limitations tables (Tables 34 and 35) is to display notable limitations identified in each study. Other notable limitations include a relatively short follow-up of 1 year which is inadequate to assess the durability of efficacy and safety (especially adverse events that are potentially rare or have delayed onset). To address the lack of evidence on long-term efficacy and safety (beyond 1 year), registries or long-term follow-up should universally enroll all patients that receive risdiplam as optional enrollment in longer follow-up studies is prone to selection bias.

Table 31. Summary of Pivotal Trial Characteristics of Risdiplam for SMA Type II and III
Study Study Type Country Dates Participants Treatment Follow-Up
SUNFISH15,51,28, DB-RCT US, EU, South America, Middle East (43 sites) 201 7-ongoing
  • Symptomatic Type 2 or 3 SMA, ambulatory or non-ambulatorya
    Age 2-25 years
  • Confirmed genetic diagnosis of 5q-autosomal recessive SMA
  • Randomization by agec
  • Primary endpoint: Change from baseline to month 12 in MFM32 score
  • Key secondary endpoint: % of patients with a ≥3- change from baseline to month 12 in the MFM32 total score

Patient characteristics (Part 2; n=180; non-ambulatory)

  • Female: 91 (50.6%)
  • Age group (years):
    • 2-5: 55 (30.6%)
    • 6-11: 57 (31.7%)
    • 12-17: 46 (25.6%)
    • 18-25: 22 (12.2%)
  • SMA type:
    • 2: 128 (71.1%)
    • 3: 52 (28.9%)
  • Mean age at onset of symptoms: 15.5 months
  • SMN2 copy number:
    • 2: 4 (2.2%)
    • 3: 157 (87.2%)
    • 4: 18 (10%)
    • Unknown: 1 (1.7%)
  • Part 1 (Dose Finding; n=51; duration 12 weeks)
  • Part 2 (double-blind randomized; n=180; all patients switched to selected treatment dose of risdiplam and followed up in open-label phase)
Intended follow-up: 12 months
DB-RCT; double-blind randomized controlled trial; EU: Europe;MFM-32: Motor Function Measure (32 items); SMA: spinal muscular atrophy.a In Part 1 ambulant or non-ambulant patients were allowed (14% were ambulatory). In Part 2, only non-ambulant patients were allowed.b Stratified by age group (2 to 5, 6 to 11, 12 to 17, or 18 to 25 years of age).
Table 32. Summary of Results of Pivotal Trial of Risdiplam for SMA Type II and III at 12 Months
Study Risdiplam (N=120) Placebo (N=60) Treatment Comparison
SUNFISH (N=180)15,51,28,      
Primary Endpoint: Total MFM-32 Scorea N=115 N=59  
Baseline, mean (SD) 45.5 (12.1) 47.4 (10.1)  
Change from baseline at month 12, LS Mean (95% CI) 1.36 (0.61 to 2.11) -0.19 (-1.22 to 0.84) 1.55 (0.30 to 2.81); p=.0156
Secondary Endpoints      
MFM-32: % of patients (95% CI) with ≥3 point change (responder analysis) 38.3% (28.9 to 47.6%) 23.7% (12.0 to 35.4%) OR (95% CI): 2.35 (1.01 to 5.44); p=.0469
RULM Total Score N=119 N=58  
Baseline, mean (SD) 19.7 (7.2) 20.9 (6.4)  
Change from baseline at month 12, LS mean (95% CI) 1.59 (0.98 to 2.20) 0.02 (-0.83 to 0.88) 1.59 (0.53 to 2.60); p=.0028
HFMSE Score      
Change from baseline at month 12, mean (SE) 0.95 (0.33) 0.37 (0.46) 0.58; p=.3015
FVCb      
Change from baseline in best % predicted value at month 12, mean (SE) -5.16% (1.40%) -3.11% (1.94%) -2.05%; p=.3804
SMAIS      
Change from baseline in the total score at month 12, LS mean (SE)
Caregiver reported
Patient reported

1.65 (0.50)
1.04 (0.65)

-0.91 (0.67)
-0.40 (0.86)

2.55; p=.0022
1.45; p=.1778
CGI-Cc
p-value (nominal, adjusted)
47.5% 40.0% 1.38; p=.3544
CI; Confidence Interval; CGI: Clinical Global Impression of Change Scale; FVC: Forced Vital Capacity; HFMSE: Expanded Hammersmith Functional Motor Scale; MFM-32: Motor Function Measure (32 items); RULM: Revised Upper Limb Module; SMA: Spinal Muscular Atrophy; SMAIS: Spinal Muscular Atrophy Independence Scale; SE: Standard error; SD: standard deviationa The MFM total score was calculated according to the user manual, expressed as a percentage of the maximum score possible for the scale (i.e., sum of the 32 item scores divided by 96 and multiplied by 100).b FVC was measured in participants over the age of 6 years. Exact number pf subset was not reported. c Proportion of patients rated by clinicians as “no change” or “improved”. The CGI-C is a single item measure of change in global health, using 7 response options: “Very much improved”, “Much improved”, “Minimally improved”, “No change”, “Minimally worse”, “Much worse”, “Very much worse”.
Table 33. Subgroup Analysis of the Pivotal SUNFISH Trial of Risdiplam for SMA Type II and III28,
Age Group Mean Treatment Difference (95% CI) in MFM32 Mean Treatment Difference (95% CI) in RULM
2-5 years (N=55) 3.03 (0.71 to 5.35) 3.34 (1.49 to 5.19)
6-11 years (N=57) 1.58 (-0.58 to 3.74) 1.07 (-0.81 to 2.95)
12-17 years (N=46) 1.04 (-1.31 to 3.39) 0.06 (-1.93 to 2.04)
18-25 years (N=22) -0.65 (-4.04 to 2.74) 1.74 (-1.06 to 4.53)
SMA Type    
Type 2 (n=124a, 125b) 1.54 (0.06 to 3.02) 1.19 (-0.03 to 2.42)
Type 3 (n=50a, n=52b) 1.49 (-0.94 to 3.93) 2.36 (0.42 to 4.29)
SMN2 Copy Number    
2 (n=4) -1.21 (-10.33 to 7.90) -0.36 (-7.84 to 7.12)
3 (n=152a, n=154b) 1.73 (0.35 to 3.10) 1.59 (0.45 to 2.72)
4 (n=17a, n=18b) 1.43 (-2.48 to 5.33) 1.70 (-1.45 to 4.86)
CI: Confidence Interval; MFM-32: Motor Function Measure (32 items); RULM: Revised Upper Limb Module; SMA: Spinal muscular atrophy.a Number reported for MFM32.b Number reported for RULM.
Table 34. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Follow-Upe
SUNFISH15,51,     1. Not standard or optimal (lack of head to head comparisons with current standard of care; however trial protocols were finalized prior to approval of nusinersen) 5. Clinical significant difference not specified (applies to RULM scores and other secondary outcomes) 1. Not sufficient duration for benefit
2. Not sufficient duration for harms
RULM: Revised Upper Limb Module; The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use.b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest.c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively.d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported.e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.
Table 35. Study Design and Conduct Limitations
Study Allocationa Blindingb Selective Reportingc Data Completenessd Powere Statisticalf
SUNFISH15,51,            
The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.a Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for selection bias.b Blinding key: 1. Not blinded to treatment assignment; 2. Not blinded outcome assessment; 3. Outcome assessed by treating physician.c Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication.d Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials).e Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference.f Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4. Comparative treatment effects not calculated.

Section Summary: Spinal Muscular Atrophy Type II and III

The evidence for use of risdiplam for pediatric or adult patients (2-25 years old) with type II or III SMA patients includes ongoing double-blind, 2-part, placebo-controlled SUNFISH study. Results of 180 patients (confirmatory study) from part 2 with 12 months follow-up are available. The least square mean change in MFM-32 from baseline was greater in patients receiving risdiplam compared with placebo with a difference of 1.55 (95% CI: 0.30 to 2.81). Responder analysis (using a threshold of increase in 3 or more points in MFM-32 as being clinically meaningful) showed that 38% of those receiving risdiplam met the criteria of achieving a clinically meaningful improvement in motor function compared to 24%. Notable limitations include a relatively short follow-up of 1 year which is inadequate to assess durability of efficacy and safety (especially adverse events that are potentially rare or have delayed onset).

Summary of Evidence

For individuals who have symptomatic type II or III SMA who receive risdiplam, the evidence includes a single ongoing double-blind, 2-part, placebo-controlled (SUNFISH) study. Relevant outcomes are overall survival, change in disease status, functional outcomes, health status measures, quality of life, and treatment-related mortality and morbidity. Pediatric or adult patients (2-25 years old) with type II or III SMA patients were enrolled in the SUNFISH study. Results of 180 patients from part 2 with 12 months follow-up are available. The least-square mean change in MFM-32 from baseline was greater in patients receiving risdiplam compared with placebo with a difference of 1.55 (95% CI: 0.30 to 2.81). Responder analysis (using a threshold of increase in 3 or more points in MFM-32 as being clinically meaningful) showed that 38% of those receiving risdiplam met the criteria of achieving a clinically meaningful improvement in motor function compared to 24%. Upper limb function measured by RULM showed a 1.6 point improvement in the risdiplam-treated group compared to no change in the placebo group. However, there is limited data to assess the long-term durability of treatment effect as well as safety-related to adverse events that are rare or have delayed onset. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Population

Reference No. 7

Policy Statement

[X] MedicallyNecessary [ ] Investigational

Supplemental Information

The purpose of the following information is to provide reference material. Inclusion does not imply endorsement or alignment with the evidence review conclusions.

Practice Guidelines and Position Statements

Guidelines or position statements will be considered for inclusion in ‘Supplemental Information' if they were issued by, or jointly by, a US professional society, an international society with US representation, or National Institute for Health and Care Excellence (NICE). Priority will be given to guidelines that are informed by a systematic review, include strength of evidence ratings, and include a description of management of conflict of interest.

National Institute for Health and Care Excellence

On July 24, 2019 the National Institute for Health and Care Excellence (NICE) issued technology appraisal guidance on nusinersen for treating spinal muscular atrophy. Nusinersen is recommended as an option for treating SMA only if people have pre-symptomatic SMA, or SMA types 1, 2 or 3 and the conditions laid out in the managed access agreement were followed. 58,

On December 16, 2021, the NICE issued technology appraisal guidance on risdiplam for treating spinal muscular atrophy. Risdiplam is recommended as an option for treating 5q spinal muscular atrophy in people 2 months and older with a clinical diagnosis of SMA types 1, 2 or 3 or with pre-symptomatic SMA and 1 to 4 SMN2 copies and the conditions laid out in the managed access agreement were followed.59,

On July 7, 2021, the NICE issued specialized technology appraisal guidance on onasemnogene abeparvovec for treating spinal muscular atrophy.60, Recommendations are summarized below.

Onasemnogene abeparvovec is recommended as an option for treating 5q spinal muscular atrophy (SMA) with a bi-allelic mutation in the SMN1 gene and a clinical diagnosis of type 1 SMA in babies, only if:

On April 19, 2023, the NICE issued a specialized technology appraisal guidance on onasemnogene abeparvovec for treating presymptomatic spinal muscular atrophy.61, Recommendations are summarized below.

Institute for Clinical and Economic Review

The Institute for Clinical and Economic Review published a final report on comparative effectiveness and value of nusinersen and onasemnogene abeparvovec-xioi for spinal muscular atrophy on April 3, 2019, and subsequently on May 24, 2019, published an update following U.S. Food and Drug Administration (FDA) approval of onasemnogene abeparvovec-xioi.38,

Nusinersen

Based on the lack of relevant data, the report concluded that the evidence for nusinersen was insufficient for type 0 and IV spinal muscular atrophy.

Onasemnogene Abeparvovec-Xioi

The report only included and appraised the published evidence from the Phase I dose-finding study of onasemnogene abeparvovec-xioi. The authors did not rate the quality of this study because they do not conduct quality assessment of non-comparative studies.

Subsequent to the FDA approval of onasemnogene abeparvovec-xioi, the Institute for Clinical and Economic Review issued an update with a brief discussion of additional data/interim analyses from ongoing trials that were presented at the Muscular Dystrophy Association Clinical and Scientific Conference April 13-17, 2019 and American Academy of Neurology Annual Meeting May 4-10, 2019) and manufacturer press releases. In summary, the Institute for Clinical and Economic Review noted that the updated data are largely consistent with previously available findings and as the data evolves and confirm the initial findings, the evidence rating may be revised.

U.S. Preventive Services Task Force Recommendations

Not applicable

Medicare National Coverage

There is no national coverage determination. In the absence of a national coverage determination, coverage decisions are left to the discretion of local Medicare carriers.

Ongoing and Unpublished Clinical Trials

Some currently unpublished and ongoing trials that might influence this review are listed in Table 33.

Table 33. Summary of Key Trials
NCT No. Trial Name Planned Enrollment Completion Date
Ongoing (Nusinersen)      
NCT04488133 (RESPOND)a A Study of Nusinersen Among Participants With Spinal Muscular Atrophy Who Received Onasemnogene Abeparvovec 46 Oct 2025
NCT04089566 (DEVOTE)a Study of Nusinersen in Participants With Spinal Muscular Atrophy 145 Jun 2024
NCT05067790 (ASCEND) A Phase 3b Study to Evaluate Higher Dose Nusinersen (BIIB058) in Patients With Spinal Muscular Atrophy Previously Treated With Risdiplam 45 Jun 2027
NCT03709784 (SAS) Spinraza in Adult Spinal Muscular Atrophy (SAS) 148 Jan 2025
NCT04729907 (ONWARD) Extension Study of Nusinersen (BIIB058) in Participants With Spinal Muscular Atrophy Who Previously Participated in a Study With Nusinersen 145 Jul 2026
Unpublished      
NCT02594124 (SHINE)a A Study for Participants With Spinal Muscular Atrophy Who Previously Participated in Nusinersen (ISIS 396443) Investigational Studies 292 Aug 2023
Ongoing (Onasemnogene abeparvovec)      
NCT03421977 (START)a Long-Term Follow-up Study for Patients From AVXS-101-CL-101 15 Dec 2033
NCT04042025a Long-term Follow-up Study of Patients Receiving Onasemnogene Abeparvovec-xioi 85 Dec 2035
NCT05386680 (STRENGTH) Phase IIIb, Open-label, Single-arm, Multi-center Study to Evaluate the Safety, Tolerability and Efficacy of OAV101 Administered Intrathecally to Participants With SMA Who Discontinued Treatment With Nusinersen or Risdiplam 28 Oct 2024
NCT05089656 (STEER) A Randomized, Sham-controlled, Double-blind Study to Evaluate the Efficacy and Safety of Intrathecal OAV101 in Type 2 Spinal Muscular Atrophy (SMA) Patients Who Are ≥ 2 to < 18 Years of Age, Treatment Naive, Sitting, and Never Ambulatory 125 Oct 2024
NCT05335876 Long-term Follow-up of Patients With Spinal Muscular Atrophy Treated With OAV101 IT or OAV101 IV in Clinical Trials 260 Oct 2039
Unpublished      
NCT03381729 (STRONG)a Study of Intrathecal Administration of Onasemnogene Abeparvovec-xioi for Spinal Muscular Atrophy 32 Nov, 2021
Ongoing (Risdiplam)      
NCT03779334 (RAINBOWFISH) A Study of Risdiplam in Infants With Genetically Diagnosed and Presymptomatic Spinal Muscular Atrophy 25 Jun 2023
NCT03032172 (JEWELFISH) A Study of Risdiplam (RO7034067) in Adult and Pediatric Participants With Spinal Muscular Atrophy 174 Jan 2022
NCT05522361 (RISE) Risdiplam Exchange in Patients With Spinal Muscular Atrophy (SMA) Previously and Exclusively Treated With Nusinersen 10 Jan 2026
NCT05115110 A Study to Investigate the Safety and Efficacy of RO7204239 in Combination With Risdiplam (RO7034067) in Ambulatory Children With Spinal Muscular Atrophy 180 Dec 2026
NCT05232929 (WeSMA) Long-Term Follow-Up Study of Patients With Spinal Muscular Atrophy Receiving Risdiplam Treatment 500 Feb 2029
Registries      
NCT04174157 Registry of Patients With a Diagnosis of Spinal Muscular Atrophy (SMA) 500 Jun 2038
NCT05042921 Pediatric SMA China Registry 300 Jun 2024
NCT05618379 Adult Spinal Muscular Atrophy (SMA) China Registry 200 Jun 2028
NCT04177134 French Register of Patients With Spinal Muscular Atrophy 1000 Jan 2029
NCT05475691 Longitudinal Data Collection in Pediatric and Adult Patients With Spinal Muscular Atrophy in Latin America 300 Jan 2025
NCT: national clinical trial; SMA: spinal muscular atrophy.a Denotes industry-sponsored or cosponsored trial.

References

  1. Prior TW, Snyder PJ, Rink BD, et al. Newborn and carrier screening for spinal muscular atrophy. Am J Med Genet A. Jul 2010; 152A(7): 1608-16. PMID 20578137
  2. Wang CH, Finkel RS, Bertini ES, et al. Consensus statement for standard of care in spinal muscular atrophy. J Child Neurol. Aug 2007; 22(8): 1027-49. PMID 17761659
  3. Lorson CL, Hahnen E, Androphy EJ, et al. A single nucleotide in the SMN gene regulates splicing and is responsible for spinal muscular atrophy. Proc Natl Acad Sci U S A. May 25 1999; 96(11): 6307-11. PMID 10339583
  4. Lefebvre S, Burlet P, Liu Q, et al. Correlation between severity and SMN protein level in spinal muscular atrophy. Nat Genet. Jul 1997; 16(3): 265-9. PMID 9207792
  5. Feldkötter M, Schwarzer V, Wirth R, et al. Quantitative analyses of SMN1 and SMN2 based on real-time lightCycler PCR: fast and highly reliable carrier testing and prediction of severity of spinal muscular atrophy. Am J Hum Genet. Feb 2002; 70(2): 358-68. PMID 11791208
  6. Muscular Dystrophy Association. Spinal Muscular Atrophy. n.d.; https://www.mda.org/disease/spinal-muscular-atrophy. Accessed August 12, 2021
  7. National Organization for Rare Disorders, Russman B. Spinal Muscular Atrophy. 2012; https://rarediseases.org/rare-diseases/spinal-muscular-atrophy/. Accessed January 14, 2024.
  8. Zerres K, Rudnik-Schöneborn S. Natural history in proximal spinal muscular atrophy. Clinical analysis of 445 patients and suggestions for a modification of existing classifications. Arch Neurol. May 1995; 52(5): 518-23. PMID 7733848
  9. Finkel RS, McDermott MP, Kaufmann P, et al. Observational study of spinal muscular atrophy type I and implications for clinical trials. Neurology. Aug 26 2014; 83(9): 810-7. PMID 25080519
  10. Rudnik-Schöneborn S, Hausmanowa-Petrusewicz I, Borkowska J, et al. The predictive value of achieved motor milestones assessed in 441 patients with infantile spinal muscular atrophy types II and III. Eur Neurol. 2001; 45(3): 174-81. PMID 11306862
  11. Farrar MA, Vucic S, Johnston HM, et al. Pathophysiological insights derived by natural history and motor function of spinal muscular atrophy. J Pediatr. Jan 2013; 162(1): 155-9. PMID 22809660
  12. Prior TW. Perspectives and diagnostic considerations in spinal muscular atrophy. Genet Med. Mar 2010; 12(3): 145-52. PMID 20057317
  13. Sugarman EA, Nagan N, Zhu H, et al. Pan-ethnic carrier screening and prenatal diagnosis for spinal muscular atrophy: clinical laboratory analysis of 72,400 specimens. Eur J Hum Genet. Jan 2012; 20(1): 27-32. PMID 21811307
  14. Biogen Inc. Highlights of Prescribing Information: Spinraza (nusinersen) injection, for intrathecal use: Prescribing label. 2016; https://www.spinraza.com/content/dam/commercial/specialty/spinraza/caregiver/en_us/pdf/spinraza-prescribing-information.pdf. Accessed January 14, 2024.
  15. Genentech. Highlights of Prescribing Information: EVRYSDI (risdiplam) for oral solution: Prescribing label. 2020; https://www.gene.com/download/pdf/evrysdi_prescribing.pdf. Accessed Accessed January 14, 2024.
  16. Albers CA, Grieve AJ. Test Review: Bayley, N. (2006). Bayley Scales of Infant and Toddler Development Third Edition. San Antonio, TX: Harcourt Assessment. J Psychoeduc Assess. 2007;25:18090
  17. Glanzman AM, Mazzone E, Main M, et al. The Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP INTEND): test development and reliability. Neuromuscul Disord. Mar 2010; 20(3): 155-61. PMID 20074952
  18. Glanzman AM, McDermott MP, Montes J, et al. Validation of the Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders (CHOP INTEND). Pediatr Phys Ther. 2011; 23(4): 322-6. PMID 22090068
  19. Finkel RS, Chiriboga CA, Vajsar J, et al. Treatment of infantile-onset spinal muscular atrophy with nusinersen: a phase 2, open-label, dose-escalation study. Lancet. Dec 17 2016; 388(10063): 3017-3026. PMID 27939059
  20. Glanzman AM, O'Hagen JM, McDermott MP, et al. Validation of the Expanded Hammersmith Functional Motor Scale in spinal muscular atrophy type II and III. J Child Neurol. Dec 2011; 26(12): 1499-507. PMID 21940700
  21. Mercuri E, Finkel R, Montes J, et al. Patterns of disease progression in type 2 and 3 SMA: Implications for clinical trials. Neuromuscul Disord. Feb 2016; 26(2): 126-31. PMID 26776503
  22. Kaufmann P, McDermott MP, Darras BT, et al. Prospective cohort study of spinal muscular atrophy types 2 and 3. Neurology. Oct 30 2012; 79(18): 1889-97. PMID 23077013
  23. De Sanctis R, Coratti G, Pasternak A, et al. Developmental milestones in type I spinal muscular atrophy. Neuromuscul Disord. Nov 2016; 26(11): 754-759. PMID 27769560
  24. Vuillerot C, Payan C, Iwaz J, et al. Responsiveness of the motor function measure in patients with spinal muscular atrophy. Arch Phys Med Rehabil. Aug 2013; 94(8): 1555-61. PMID 23380348
  25. Trundell D, Le Scouiller S, Gorni K, et al. Validity and Reliability of the 32-Item Motor Function Measure in 2- to 5-Year-Olds with Neuromuscular Disorders and 2- to 25-Year-Olds with Spinal Muscular Atrophy. Neurol Ther. Dec 2020; 9(2): 575-584. PMID 32856191
  26. Trundell D, Le Scouiller S, Le Goff L, et al. Assessment of the validity and reliability of the 32-item Motor Function Measure in individuals with Type 2 or non-ambulant Type 3 spinal muscular atrophy. PLoS One. 2020; 15(9): e0238786. PMID 32946459
  27. Trundell D, Le Scouiller S, Staunton H, Gorni K and Vuillerot C. Validity and reliability of the Motor Function Measure (MFM32) in children with neuromuscular disorders (NMDs) and in individuals with Type 2 and non-ambulant Type 3 spinal muscular atrophy (SMA). Poster presented as at the Cure SMA Researcher Meeting, 23rd International SMA Research Meeting, Disneyland, Anaheim, CA, USA, June 28July 1, 2019.
  28. Center for Drug Evaluation and Research Application Number: 213535orig1s000. Clinical Review(S). Available at https://www.accessdata.fda.gov/drugsatfda_docs/nda/2020/213535Orig1s000MedR.pdf. Accessed January 14, 2024.
  29. Haataja L, Mercuri E, Regev R, et al. Optimality score for the neurologic examination of the infant at 12 and 18 months of age. J Pediatr. Aug 1999; 135(2 Pt 1): 153-61. PMID 10431108
  30. Kolb SJ, Coffey CS, Yankey JW, et al. Natural history of infantile-onset spinal muscular atrophy. Ann Neurol. Dec 2017; 82(6): 883-891. PMID 29149772
  31. Crawford TO, Swoboda KJ, De Vivo DC, et al. Continued benefit of nusinersen initiated in the presymptomatic stage of spinal muscular atrophy: 5-year update of the NURTURE study. Muscle Nerve. Aug 2023; 68(2): 157-170. PMID 37409780
  32. De Vivo DC, Bertini E, Swoboda KJ, et al. Nusinersen initiated in infants during the presymptomatic stage of spinal muscular atrophy: Interim efficacy and safety results from the Phase 2 NURTURE study. Neuromuscul Disord. Nov 2019; 29(11): 842-856. PMID 31704158
  33. Finkel RS, Mercuri E, Darras BT, et al. Nusinersen versus Sham Control in Infantile-Onset Spinal Muscular Atrophy. N Engl J Med. Nov 02 2017; 377(18): 1723-1732. PMID 29091570
  34. Acsadi G, Crawford TO, Müller-Felber W, et al. Safety and efficacy of nusinersen in spinal muscular atrophy: The EMBRACE study. Muscle Nerve. May 2021; 63(5): 668-677. PMID 33501671
  35. Chiriboga CA, Swoboda KJ, Darras BT, et al. Results from a phase 1 study of nusinersen (ISIS-SMN(Rx)) in children with spinal muscular atrophy. Neurology. Mar 08 2016; 86(10): 890-7. PMID 26865511
  36. Darras BT, Chiriboga CA, Iannaccone ST, et al. Nusinersen in later-onset spinal muscular atrophy: Long-term results from the phase 1/2 studies. Neurology. May 21 2019; 92(21): e2492-e2506. PMID 31019106
  37. Mercuri E, Darras BT, Chiriboga CA, et al. Nusinersen versus Sham Control in Later-Onset Spinal Muscular Atrophy. N Engl J Med. Feb 15 2018; 378(7): 625-635. PMID 29443664
  38. Institute for Clinical and Evidence Review. Spinraza and Zolgensma for Spinal Muscular Atrophy: Effectiveness and Value (Final Evidence Report April 3, 2019; Updated May 24, 2019). 2019; https://icer-review.org/wp-content/uploads/2018/07/ICER_SMA_Final_Evidence_Report_052419.pdf. Accessed January 14, 2024.
  39. Strauss KA, Farrar MA, Muntoni F, et al. Onasemnogene abeparvovec for presymptomatic infants with two copies of SMN2 at risk for spinal muscular atrophy type 1: the Phase III SPR1NT trial. Nat Med. Jul 2022; 28(7): 1381-1389. PMID 35715566
  40. Strauss KA, Farrar MA, Muntoni F, et al. Onasemnogene abeparvovec for presymptomatic infants with three copies of SMN2 at risk for spinal muscular atrophy: the Phase III SPR1NT trial. Nat Med. Jul 2022; 28(7): 1390-1397. PMID 35715567
  41. Mendell JR, Al-Zaidy S, Shell R, et al. Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med. Nov 02 2017; 377(18): 1713-1722. PMID 29091557
  42. Day JW, Finkel RS, Chiriboga CA, et al. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy in patients with two copies of SMN2 (STR1VE): an open-label, single-arm, multicentre, phase 3 trial. Lancet Neurol. Apr 2021; 20(4): 284-293. PMID 33743238
  43. Mercuri E, Muntoni F, Baranello G, et al. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy type 1 (STR1VE-EU): an open-label, single-arm, multicentre, phase 3 trial. Lancet Neurol. Oct 2021; 20(10): 832-841. PMID 34536405
  44. Mendell JR, Al-Zaidy SA, Lehman KJ, et al. Five-Year Extension Results of the Phase 1 START Trial of Onasemnogene Abeparvovec in Spinal Muscular Atrophy. JAMA Neurol. Jul 01 2021; 78(7): 834-841. PMID 33999158
  45. Finkel RS, Darras BT, Mendell JR, et al. Intrathecal Onasemnogene Abeparvovec for Sitting, Nonambulatory Patients with Spinal Muscular Atrophy: Phase I Ascending-Dose Study (STRONG). J Neuromuscul Dis. 2023; 10(3): 389-404. PMID 36911944
  46. Finkel RS, Day JW, De Vivo DC, et al. RESTORE: A Prospective Multinational Registry of Patients with Genetically Confirmed Spinal Muscular Atrophy - Rationale and Study Design. J Neuromuscul Dis. 2020; 7(2): 145-152. PMID 32039859
  47. Prior TW, Krainer AR, Hua Y, et al. A positive modifier of spinal muscular atrophy in the SMN2 gene. Am J Hum Genet. Sep 2009; 85(3): 408-13. PMID 19716110
  48. Novartis Inc. A. Highlights of Prescribing Information: Zolgensma (onasemnogene abeparvovec-xioi) suspension for intravenous infusion: Prescribing label. 2019; https://www.novartis.com/us-en/sites/novartis_us/files/zolgensma.pdf. Accessed January 14, 2024.
  49. Lin CW, Kalb SJ, Yeh WS. Delay in Diagnosis of Spinal Muscular Atrophy: A Systematic Literature Review. Pediatr Neurol. Oct 2015; 53(4): 293-300. PMID 26260993
  50. WHO Multicentre Growth Reference Study Group. WHO Motor Development Study: windows of achievement for six gross motor development milestones. Acta Paediatr Suppl. Apr 2006; 450: 86-95. PMID 16817682
  51. Darras BT, Masson R, Mazurkiewicz-Bełdzińska M, et al. Risdiplam-Treated Infants with Type 1 Spinal Muscular Atrophy versus Historical Controls. N Engl J Med. Jul 29 2021; 385(5): 427-435. PMID 34320287
  52. Masson R, Mazurkiewicz-Bełdzińska M, Rose K, et al. Safety and efficacy of risdiplam in patients with type 1 spinal muscular atrophy (FIREFISH part 2): secondary analyses from an open-label trial. Lancet Neurol. Dec 2022; 21(12): 1110-1119. PMID 36244364
  53. Baranello G, Darras BT, Day JW, et al. Risdiplam in Type 1 Spinal Muscular Atrophy. N Engl J Med. Mar 11 2021; 384(10): 915-923. PMID 33626251
  54. Mercuri E, Deconinck N, Mazzone ES, et al. Safety and efficacy of once-daily risdiplam in type 2 and non-ambulant type 3 spinal muscular atrophy (SUNFISH part 2): a phase 3, double-blind, randomised, placebo-controlled trial. Lancet Neurol. Jan 2022; 21(1): 42-52. PMID 34942136
  55. Oskoui M, Day JW, Deconinck N, et al. Two-year efficacy and safety of risdiplam in patients with type 2 or non-ambulant type 3 spinal muscular atrophy (SMA). J Neurol. May 2023; 270(5): 2531-2546. PMID 36735057
  56. Chiriboga CA, Bruno C, Duong T, et al. Risdiplam in Patients Previously Treated with Other Therapies for Spinal Muscular Atrophy: An Interim Analysis from the JEWELFISH Study. Neurol Ther. Apr 2023; 12(2): 543-557. PMID 36780114
  57. Center for Drug Evaluation and Research Application Number: 213535orig1s000. Clinical Review(S). Available at https://www.accessdata.fda.gov/drugsatfda_docs/nda/2020/213535Orig1s000MedR.pdf. Accessed January 14, 2024.
  58. Nusinersen for treating spinal muscular atrophy. Published July 24, 2019. Available at https://www.nice.org.uk/guidance/ta588. Accessed on January 14, 2024.
  59. Risdiplam for treating spinal muscular atrophy: Technology appraisal guidance. Published: 16 December 2021. Available at www.nice.org.uk/guidance/ta755. Accessed on January 14, 2024.
  60. Onasemnogene abeparvovec for treating spinal muscular atrophy. Highly specialised technologies guidance. Published: 7 July 2021. Available at www.nice.org.uk/guidance/hst15. Accessed on January 14, 2024.
  61. Onasemnogene abeparvovec for treating presymptomatic spinal muscular atrophy. Highly specialised technologies guidance. Published: 19 April 2023. Available at www.nice.org.uk/guidance/hst24. Accessed on January 14, 2024.

Codes

Codes Number Description
CPT 96450 Chemotherapy administration, into CNS (eg, intrathecal), requiring and including spinal puncture
  0236U SMN1 (survival of motor neuron 1, telomeric) and SMN2 (survival of motor neuron 2, centromeric) (eg, spinal muscular atrophy) full gene analysis, including small sequence changes in exonic and intronic regions, duplications and deletions, and mobile element insertions
HCPCS J2326 Injection, nusinersen, 0.1 mg
  J3399 Injection, onasemnogene abeparvovec-xioi, per treatment, up to 5x10^15 vector genomes
  J8499 No specific code use for: Evrysdi (risdiplam) for oral solution
ICD-10-CM G12.0 Infantile spinal muscular atrophy, type I [Werdnig-Hoffman]
  G12.1 Other inherited spinal muscular atrophy
  Z92.86 Personal history of gene therapy
ICD-10-PCS   ICD-10-PCS codes are only used for inpatient services.
  3E0R3GC Administration, physiological systems and anatomical regions, introduction, spinal canal, percutaneous, other therapeutic substance
Type of Service Medicine  
Place of Service Inpatient/Outpatient

Policy History

Date Action Description
04/19/2024 Review due to MPP Policy updated with literature review through January 8, 2024; references added. Policy statements edited to improve clarity.
08/16/2023  Annual Review Policy updated with literature review through January 6, 2023; multiple references were added. The conclusions for sufficiency of evidence to determine that the technology results in an improvement in the net health outcome was changed from insufficient to sufficient for individuals who are presymptomatic with a genetic diagnosis of spinal muscular atrophy and less than 4 copies of SMN2 who receive onasemnogene abeparvovec-xioi.Policy statement updated to remove "Diagnosis of SMA based on the results of SMA newborn screening" and added the verbigage to the policy guidelines section. Removed onasemnogene abeparvovec-xioi high-control leaving only one statement for onasemnogene abeparvovec-xioi.
08/23/2022  Policy Review Policy governance (review and vote for approval) transferred to National Pharmacy and Therapeutics Committee in March 2022. Policy statements for nusinersen, onasemnogene abeparvovec-xioi and risdiplam were modified. High-control and low-control versions of policy statements for onasemnogene abeparvovec-xioi were added. Policy statements for risdiplam were modified to include infants with spinal muscular atrophy less than 2 months of age based on FDA approved label extension.
11/03/2021  Annual Review Policy updated with literature review through August 12, 2021; references added. Policy statements unchanged.
11/10/2020  Policy Review Policy updated with literature review through August 12, 2020; references added. Policy statements for nusinersen were modified. Rationale and policy statements added for risdiplam.
05/29/2020  Annual Review Policy updated with literature review through March 1, 2020; reference added. Policy statements unchanged.
07/15/2019  Policy Review Policy updated with literature review through May 24, 2019; relevant information on onasemnogene abeparvovec-xioi was added. Policy statements for nusinersen were modified. Policy statements and rationale for onasemnogene abeparvovec-xioi were added.
04/12/2018 New Policy – Add to Prescription Drugs section Policy created