Medical Policy

Policy Num:       05.001.048
Policy Name:     Biological Treatments for Refractory Myasthenia Gravis
Policy ID:           [05.001.048]  [Ac / B / M+ / P+]  [5.01.39]


Last Review:      July 17, 2024
Next Review:      July 20, 2025

 

Related Policies: None

Biological Treatments for Refractory Myasthenia Gravis

Population Reference No. Populations Interventions Comparators Outcomes

1

Individuals:
  • Adults with generalized myasthenia gravis who are anti-acetylcholine receptor antibody positive
Interventions of interest are:
  • Eculizumab
Comparators of interest are:
  • Standard of care
Relevant outcomes include:
  • Symptoms
  • Quality of life
  • Hospitalizations
  • Resource utilization

2

Individuals:
  • Adults with generalized myasthenia gravis who are anti-acetylcholine receptor antibody positive
Interventions of interest are:
  • Ravulizumab-cwvz
Comparators of interest are:
  • Standard of care
Relevant outcomes include:
  • Symptoms
  • Quality of life
  • Hospitalizations
  • Resource utilization

3

Individuals:
  • Adults with generalized myasthenia gravis who are anti-acetylcholine receptor antibody positive
Interventions of interest are:
  • Efgartigimod alfa-fcab (intravenous) or efgartigimod alfa and hyaluronidase-qvfc(subcutaneous)
Comparators of interest are:
  • Standard of care
Relevant outcomes include:
  • Symptoms
  • Quality of life
  • Hospitalizations
  • Resource utilization

4

Individuals:
  • Adults with generalized myasthenia gravis who are anti-acetylcholine receptor or antimuscle-specific tyrosine kinase antibody positive
  • Rozanolixizumab-noli
Comparators of interest are:
  • Standard of care
Relevant outcomes include:
  • Symptoms
  • Quality of life
  • Hospitalizations
  • Resource utilization

Summary

Summary

Description

Myasthenia gravis is an autoimmune neuromuscular disorder characterized by fluctuating motor weakness involving ocular, bulbar, limb, and/or respiratory muscles. The weakness is due to an antibody-mediated, immunologic attack directed at proteins in the postsynaptic membrane of the neuromuscular junction (acetylcholine receptors or receptor-associated proteins). Eighty to 90 percent of individuals with myasthenia gravis have autoantibodies against the acetylcholine receptor detectable in serum, and these antibodies are believed to play a central role in disease pathomechanism. Eculizumab (Soliris®) and ravulizumab-cwvz (Ultomiris®) are monoclonal antibodies that are presumed to exert a therapeutic effect in individuals with generalized myasthenia gravis through the reduction of terminal complement complex C5b-9 deposition at the neuromuscular junction. Efgartigimod alfa-fcab (Vyvgart®) is a human IgG1 antibody fragment that binds to the neonatal Fc receptor, resulting in the reduction of circulating IgG in individuals with generalized myasthenia gravis. Vygart Hytrulo is a coformulation of efgartigimod alfa and hyaluronidase (human recombinant) which can be administered subcutaneously. The addition of hyaluronidase increases the dispersion and absorption of co-administered drugs when administered subcutaneously. Rozanolixizumab-noli is a humanized IgG4 monoclonal antibody that binds to the neonatal Fc receptor resulting in the reduction of circulating IgG.

Summary of Evidence

For individuals with generalized myasthenia gravis who receive eculizumab, the evidence includes a single pivotal randomized controlled trial (RCT). Relevant outcomes are symptoms, quality of life, hospitalizations, and resource utilization. Results of the pivotal REGAIN trial reported a statistically significant difference in the primary endpoint favoring eculizumab in Myasthenia Gravis-Specific Activities of Daily Living scale (MG-ADL) total scores compared with the placebo (least square mean difference of -1.9 points; 95% confidence interval [CI]: -3.3 to -0.6). A key secondary endpoint of change from baseline in the Quantitative Myasthenia Gravis (QMG) total score at week 26 also favored eculizumab compared with placebo (least square mean difference of -3.0 points; 95% CI: -4.6 to -1.3). Proportion of responders as defined by at least a 3-point reduction in MG‐ADL total score and at least a 5-point reduction in QMG total score from baseline to week 26 in eculizumab compared to placebo was 60% versus 40% and 45% versus 19%, respectively. An open-label extension of the pivotal trial provided additional evidence that showed that individuals who had received placebo during the REGAIN double-blind phase experienced rapid and sustained improvements during open-label eculizumab phase. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with generalized myasthenia gravis who receive ravulizumab, the evidence includes a single pivotal RCT. Relevant outcomes are symptoms, quality of life, hospitalizations, and resource utilization. Results of the pivotal CHAMPION MG trial reported a statistically significant difference in the primary endpoint favoring ravulizumab in MG-ADL total scores compared with the placebo (least square mean difference of -1.6 points; 95% CI: -2.6 to -0.7). A key secondary endpoint of change from baseline in the QMG total score at week 26 also favored ravulizumab compared with placebo (least square mean difference of -2.0 points; 95% CI: -3.2 to -0.8). Proportion of responders as defined by at least a 3-point reduction in MG‐ADL total score and at least a 5-point reduction in QMG total score from baseline to week 26 in eculizumab compared to placebo was 57% versus 34% and 30% versus 11%, respectively. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with generalized myasthenia gravis who receive efgartigimod, the evidence includes 2 pivotal RCTs. Relevant outcomes are symptoms, quality of life, hospitalizations, and resource utilization. Two formulations are currently approved by the Food and Drug Administration (FDA): Vygart (efgartigimod alfa-fcab injection for intravenous infusion) and Vygart Hytrulo (efgartigimod alfa and hyaluronidase-qvfc for subcutaneous use). Initial FDA approval of the intravenous formulation was based on a single RCT called ADAPT. Results of this trial reported a statistically significant difference in the primary endpoint favoring efgartigimod in MG-ADL responder rate compared with the placebo (67.7% versus 29.7%, respectively; p<.0001). MG-ADL responder was defined as a patient with a 2-point or greater reduction in the total MG-ADL score compared to the treatment cycle baseline for at least 4 consecutive weeks, with the first reduction occurring no later than 1 week after the last infusion of the cycle. A key secondary endpoint of responder based on QMG total score at week 26 also favored efgartigimod compared with placebo (63.1% versus 14.1%, respectively; p<.0001). QMG responder was defined as a patient who had a 3-point or greater reduction in the total QMG score compared to the treatment cycle baseline for at least 4 consecutive weeks, with the first reduction occurring no later than 1 week after the last infusion of the cycle. Subsequent approval of the subcutaneous formulation was based on the results of a bridging 10-week open-label randomized trial called ADAPT-SC. Results of this trial demonstrated pharmacodynamic non-inferiority based on the percent reduction in AChR-Ab levels from baseline to day 29. The least squares mean difference was 2.5% (95% CI: -7.45 to 2.41), which was below the upper limit of the confidence interval of 10%. Because of the relatively short follow-up, there is still considerable uncertainty about the long-term net benefits of efgartigimod compared with other treatment options. No major limitations in the study design and conduct were identified. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with generalized myasthenia gravis who receive rozanolixizumab, the evidence includes a single pivotal RCT. Relevant outcomes are symptoms, quality of life, hospitalizations, and resource utilization. Results of the pivotal RCT MycarinG reported a statistically significant difference in the primary endpoint favoring rozanolixizumab in MG-ADL total scores compared with the placebo (LS mean difference of -2.6 points for either dose of rozanolixizumab). A key secondary endpoint of change from baseline in the QMG total score at week 43 also favored rozanolixizumab [-5.4 points and -6.7 points in the rozanolixizumab-treated group at 7 mg/kg and 10 mg/kg dose level, respectively, compared with -1.9 points in the placebo-treated group (p<.001)]. Because of the relatively short follow-up, there is still considerable uncertainty about the long-term net benefits of rozanolixizumab compared with other treatment options. 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 with biologicals improves the net health outcome in individuals with generalized myasthenia gravis.

POLICY statements

Eculizumab and Ravulizumab-cwvz - Initial Treatment

Eculizumab and ravulizumab-cwvz may be considered medically necessary for individuals with generalized myasthenia gravis (gMG) if they meet criteria 1 through 6:

  1. 18 years of age or older.

  2. Diagnosis of gMG with a class II to IV disease per the Myasthenia Gravis Foundation of America (MGFA) classification system (see Policy Guidelines).

  3. Anti-acetylcholine receptor (AChR) antibody positive.

  4. Impaired activities of daily living defined as a MG-Activities of Daily Living (MG-ADL) total score of ≥6.

  5. Inadequate treatment response, intolerance, or contraindication to an acetylcholinesterase inhibitor (e.g., pyridostigmine, neostigmine) and at least ONE immunosuppressive therapy (e.g., azathioprine, cyclosporine, mycophenolate mofetil, tacrolimus, methotrexate, cyclophosphamide) either in combination or as monotherapy.

  6. Not receiving dual therapy with another C5 complement inhibitor for gMG.

  7. Vaccination against Neisseria meningitidis at least 2 weeks prior to initiation of therapy [unless treatment cannot be delayed].

  8. Prescribing physician is enrolled in the appropriate Risk Evaluation and Mitigation Strategies (REMS) program.

Initial authorization period is for 6 months.

Eculizumab and Ravulizumab-cwvz - Continuation of Treatment

Incremental reauthorization for eculizumab and ravulizumab-cwvz may be considered medically necessary for individuals with gMG if they meet criteria 1 through 2:

  1. Continues to meet the initial treatment criteria cited above.

  2. Decrease of 2 points in MG-ADL total score from pre-treatment baseline value.

Reauthorization period is for 12 months.

Eculizumab and ravulizumab-cwvz are considered investigational for generalized myasthenia gravis when the above criteria are not met.

Efgartigimod alfa-fcab (intravenous) and efgartigimod alfa and hyaluronidase-gvfc (subcutaneous) - Initial Treatment

Efgartigimod alfa-fcab and efgartigimod alfa and hyaluronidase-gvfc may be considered medically necessary for individuals with gMG if they meet criteria 1 through 6:

  1. 18 years of age or older.

  2. Diagnosis of gMG with class II to IV disease per the MGFA classification system (see Policy Guidelines).

  3. Anti-AChR antibody positive.

  4. Impaired activities of daily living defined as a MG-ADL total score of ≥5.

  5. Inadequate treatment response, intolerance, or contraindication to an acetylcholinesterase inhibitor (e.g., pyridostigmine, neostigmine) and at least ONE immunosuppressive therapy (e.g., azathioprine, cyclosporine, mycophenolate mofetil, tacrolimus, methotrexate, cyclophosphamide) either in combination or as monotherapy.

  6. IgG levels ≥6 g/L.

Initial authorization period is for 6 months.

Efgartigimod alfa-fcab (intravenous) and efgartigimod alfa and hyaluronidase-gvfc (subcutaneous) - Continuation of Treatment

Incremental reauthorization for efgartigimod alfa-fcab and efgartigimod alfa and hyaluronidase-gvfc (subcutaneous) may be considered medically necessary for individuals with gMG if they meet criteria 1 through 2:

  1. Continues to meet the initial treatment criteria cited above.

  2. Decrease of 2 points in MG-ADL total score from pre-treatment baseline value.

Reauthorization period is for 12 months.

Efgartigimod alfa-fcab is considered investigational when the above criteria are not met.

Rozanolixizumab-noli - Initial Treatment

Rozanolixizumab-noli may be considered medically necessary for individuals with gMG if they meet criteria 1 through 6:

  1. 18 years of age or older.

  2. Diagnosis of gMG with class II to IVa disease per the MGFA classification system (see Policy Guidelines).

  3. Anti-AChR antibody positive or anti-muscle-specific tyrosine kinase (MuSK) antibody positive.

  4. Impaired activities of daily living defined as a MG-DLS total score of ≥3 and at least 3 points from non-ocular symptom(s).

  5. Meets any one of the following:

    1. If anti-AChR antibody positive: inadequate treatment response, intolerance, or contraindication to an acetylcholinesterase inhibitor (e.g., pyridostigmine, neostigmine) and at least one immunosuppressive therapy (e.g., azathioprine, cyclosporine, mycophenolate mofetil, tacrolimus, methotrexate, cyclophosphamide) either in combination or as monotherapy.

    2. If anti-MuSK antibody positive: inadequate treatment response, intolerance, or contraindication to at least one immunosuppressive therapy (e.g., azathioprine, cyclosporine, mycophenolate mofetil, tacrolimus, methotrexate, cyclophosphamide).

  6. IgG levels ≥5.5 g/L.

Initial authorization period is for 6 months.

Rozanolixizumab-noli - Continuation of Treatment

Incremental reauthorization of rozanolixizumab-noli may be considered medically necessary for individuals with gMG if they meet criteria 1 through 2:

  1. Individual continues to meet the initial treatment criteria cited above.

  2. Decrease of 2 points in MG-ADL total score from pre-treatment baseline value.

Reauthorization period is for 12 months.

Rozanolixizumab-noli is considered investigational when the above criteria are not met.

POLICY GUIDELINES

Eculizumab

Eculizumab is supplied in a 300 mg/30 mL single-dose vial. The recommended dose of eculizumab for adults with generalized myasthenia gravis (gMG) who are anti-acetylcholine receptor (AChR) antibody positive is 900 mg IV weekly for the first 4 weeks, followed by 1200 mg for the fifth dose 1 week later, than 1200 mg every 2 weeks thereafter. Administer eculizumab at the recommended dosage regimen time points, or within 2 days of these time points. Only administer as an IV infusion. Do not administer as an IV push or bolus injection. The IV infusion should be administered over 35 minutes in adults. If an adverse reaction occurs during eculizumab administration, the infusion may be slowed or stopped at the discretion of the clinician. If the infusion is slowed, the total infusion time should not exceed 2 hours in adults. Monitor the individual for at least 1 hour following completion of the infusion for signs or symptoms of an infusion-related reaction.

Ravulizumab-cwvz

Ravulizumab-cwvz is supplied in 300 mg/30 mL, 300 mg/3 mL, and 1100 mg/11 mL single-dose vials. The recommended IV dosage regimen of ravulizumab-cwvz for adults with gMG who are anti-AChR antibody positive is weight-based as follows:

The dosing schedule is allowed to occasionally vary within 7 days of the scheduled infusion day (except for the first maintenance dose); but subsequent doses should be administered according to the original schedule. Only administer as an IV infusion through a 0.2 or 0.22 micron filter. The subcutaneous dosage form is not indicated for generalized myasthenia gravis. If an adverse reaction occurs during ravulizumab-cwvz administration, the infusion may be slowed or stopped at the discretion of the clinician. Monitor the individual for at least 1 hour following completion of the infusion for signs or symptoms of an infusion-related reaction.

Efgartigimod alfa-fcab

Efgartigimod is supplied in a 400 mg/20 mL single-dose vial. The recommended dose of efgartigimod for adults with gMG who are anti-AChR antibody positive is 10 mg/kg given as an IV infusion over 1 hour once weekly for 4 weeks. In individuals weighing ≥120 kg, the recommended dose is 1200 mg per infusion. Providers should avoid administration of efgartigimod alfa-fcab to individuals with an active infection. Administer subsequent treatment cycles based on clinical evaluation. The safety of initiating subsequent cycles sooner than 50 days from the start of the previous treatment cycle has not been established. If a scheduled infusion is missed, the drug may be given up to 3 days after the scheduled time point. Thereafter, resume the original dosing schedule until the treatment cycle is completed. Monitor individuals during administration and for 1 hour thereafter for clinical signs and symptoms of hypersensitivity reactions. If such a reaction occurs, discontinue therapy and institute appropriate supportive measures. Providers should evaluate the need to administer age-appropriate vaccines according to immunization guidelines before initiation of a new treatment cycle with efgartigimod alfa-fcab.

Efgartigimod alfa and hyaluronidase-gvfc

Efgartigimod alfa and hyaluronidase-gvfc is supplied as a single-dose vial containing 1008 mg efgartigimod alfa and 11,200 units hyaluronidase per 5.6 mL. Efgartigimod alfa and hyaluronidase-gvfc is intended for subcutaneous administration every 4 weeks by a healthcare professional. Administer subsequent treatment cycles based on clinical evaluation. The safety of initiating subsequent cycles sooner than 50 days from the start of the previous treatment cycle has not been established. If a scheduled infusion is missed, the drug may be given up to 3 days after the scheduled time point. Thereafter, resume the original dosing schedule until the treatment cycle is completed. Monitor individuals during administration and for 30 minutes thereafter for clinical signs and symptoms of hypersensitivity reactions. If such a reaction occurs, discontinue therapy and institute appropriate supportive measures. Prescribers should evaluate the need to administer age-appropriate vaccines according to immunization guidelines before initiation of a new treatment cycle with efgartigimod alfa and hyaluronidase-gvfc.

Rozanolixizumab-noli

Rozanolixizumab-noli is supplied single-dose glass vial containing 280 mg/2 mL. The recommended dosage regimen of rozanolixizumab-noli is weight-based as follows:

Administer subsequent treatment cycles based on clinical evaluation. The safety of initiating subsequent cycles sooner than 63 days from the start of the previous treatment cycle has not been established. If a scheduled infusion is missed, the drug may be given up to 4 days after the scheduled time point. Thereafter, resume the original dosing schedule until the treatment cycle is completed.

It is recommended that individuals be monitored during administration and for 15 minutes thereafter for clinical signs and symptoms of hypersensitivity reactions, including angioedema and rash. If such a reaction occurs, discontinue therapy and institute appropriate supportive measures. In clinical trials, hypersensitivity reactions occurred up to 2 weeks post-administration.

It is recommended that individuals are monitored for symptoms consistent with aseptic meningitis. In clinical trials, a total of 3 individuals developed drug-inducced aseptic meningitis, which lead to hospitalization and discontinuation of rozanolixizumab-noli.

Boxed Warning - Eculizumab and Ravulizumab-cwvz

There is a boxed warning regarding the potential for serious life-threatening and fatal meningococcal infections. The warning recommends that clinicians:

Advisory Committee on Immunization Practices Recommendations

Recent data suggest that meningococcal vaccines likely provide incomplete protection against invasive meningococcal disease in individuals receiving eculizumab. Experts believe this increased risk likely also applies to individuals receiving ravulizumab-cwvz. The Advisory Committee on Immunization Practices (ACIP) recommends both the meningococcal conjugate (MenACWY) and serogroup B meningococcal (MenB) vaccines for individuals receiving these complement inhibitors. Depending on the brand, the full series of MenB vaccine requires 2 or 3 doses, Administer a booster dose of MenACWY vaccine every 5 years, for the duration of complement inhibitor therapy. Administer a booster dose of MenB vaccine 1 year after series completion and then every 2 to 3 years thereafter, for the duration of therapy.

Risk Evaluation and Mitigation Strategies - Eculizumab and Ravulizumab-cwvz

The Food and Drug Administration (FDA) approved eculizumab and ravulizumab-cwvz with risk evaluation and mitigation strategies (REMS) in order to mitigate the occurrence and morbidity associated with meningococcal infections. Clinicians who prescribe eculizumab and ravulizumab-cwvz must:

Myasthenia Gravis Foundation of America Clinical Classification

In 1997, the Medical Scientific Advisory Board of the Myasthenia Gravis Foundation of America (MGFA) formed a task force to address the need for universally accepted classifications, grading systems, and analytic methods for management of individuals undergoing therapy and for use in therapeutic research trials. As a result, the MGFA Clinical Classification was created. This classification divides myasthenia gravis (MG) into 5 main classes and several subclasses, as follows:

Class I: Any ocular muscle weakness; may have weakness of eye closure. All other muscle strength is normal.

Class II: Mild weakness affecting muscles other than ocular muscles; may also have ocular muscle weakness of any severity.

IIa. Predominantly affecting limb, axial muscles, or both. May also have lesser involvement of oropharyngeal muscles.

IIb. Predominantly affecting oropharyngeal, respiratory muscles, or both. May also have lesser or equal involvement of limb, axial muscles, or both.

Class III: Moderate weakness affecting muscles other than ocular muscles; may also have ocular muscle weakness of any severity.

IIIa. Predominantly affecting limb, axial muscles, or both. May also have lesser involvement of oropharyngeal muscles.

IIIb. Predominantly affecting oropharyngeal, respiratory muscles, or both. May also have lesser or equal involvement of limb, axial muscles, or both.

Class IV: Severe weakness affecting muscles other than ocular muscles; may also have ocular muscle weakness of any severity.

IVa. Predominantly affecting limb, axial muscles, or both. May also have lesser involvement of oropharyngeal muscles.

IVb. Predominantly affecting oropharyngeal, respiratory muscles, or both. May also have lesser or equal involvement of limb, axial muscles, or both.

Class V: Defined as intubation, with or without mechanical ventilation, except when employed during routine postoperative management. The use of a feeding tube without intubation places the individual in class IVb.

Coding

See the Codes table for details.

BENEFIT APPLICATION

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.

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.

BACKGROUND

Myasthenia Gravis

Myasthenia gravis is an acquired, autoimmune disorder that affects the neuromuscular junction of the skeletal muscles. Eighty to 90 percent of individuals with myasthenia gravis have autoantibodies against the acetylcholine receptor (AChR) detectable in serum, and these antibodies are believed to play a central role in disease pathomechanism. The AChR antibodies in myasthenia gravis are primarily immunoglobulin G1 (IgG1) and G3 (IgG3). In addition to blocking ACh binding to the AChR and cross-linking and internalizing the AChRs, these antibodies act through complement activation.1, Some individuals with myasthenia gravis who are seronegative for AChR antibodies have antibodies directed against another target on the surface of the muscle membrane, muscle-specific receptor tyrosine kinase.2, In contrast with AChR antibody-positive myasthenia gravis, in which complement-fixing immunoglobulin G1 (IgG1) and G3 (IgG3) subclasses predominate3,, muscle-specific kinase antibodies are mainly IgG44,, the IgG subtype that does not activate complement.

The clinical manifestations can vary from mild and focal weakness in some individuals to severe tetraparesis with respiratory failure in others. Symptom severity may also vary substantially in an individual patient throughout the day and over the course of the condition. Classification systems stratify individuals by symptoms or diagnostic findings to specify the severity of impairment and to aid with management. There are 2 clinical forms - ocular and generalized. In ocular from, weakness is limited to the eyelids and extraocular muscles while in generalized form, weakness involves a variable combination of ocular, bulbar, limb, and respiratory muscles. Myasthenia gravis may be categorized by symptom severity to guide treatment decisions, determine eligibility for clinical trials, and help with prognostication. A widely used classification system from a task force of the Myasthenia Gravis Foundation of America stratifies individuals by the extent and severity of muscle weakness5, and is summarized in the section of "Policy Guidelines". Myasthenia gravis is a relatively uncommon disorder. Both incidence and prevalence have significant geographical variations. Reported prevalence rates range from 150 to 200 cases per million, and they have steadily increased over the past 50 years, at least partly due to improvements in recognition, diagnosis, treatment, and an overall increase in life expectancy.6, More recent studies addressing incidence rates have been conducted in Europe and show a wide range from 4.1 to 30 cases per million person-years.7,8, The annual rate is lower in studies coming from North America and Japan, with the incidence ranging from 3 to 9.1 cases per million.9,

The diagnosis is primarily based on clinical testing. Laboratory investigations and procedures can aid the clinician in confirming clinical findings. These may include serologic tests, electrophysiologic exams (eg, repetitive nerve stimulation test and single-fiber electromyography), an edrophonium test, an ice-pack test, imaging, and laboratory testing for other coexisting autoimmune disorders (eg, anti-nuclear antibodies, rheumatoid factor, and thyroid function). For most individuals with clinical features of myasthenia gravis, the diagnosis is confirmed by the presence of autoantibodies against the AChRs or against other muscle receptor-associated proteins. A positive anti-AChR antibody is present in 80% of individuals with gMG and confirms the diagnosis in an individual with classical clinical findings. About 5 to 10% of individuals will demonstrate anti-muscle specific kinase antibodies. Individuals who are seronegative for either of these antibodies will have anti-LRP4 antibodies.

Treatment

The goals of therapy are to render individuals minimally symptomatic or better while minimizing side effects from medications. The 4 basic therapies for myasthenia gravis include: 1) symptomatic therapy with an acetylcholinesterase inhibitor such as pyridostigmine and neostigmine; 2) chronic immunotherapies (such as glucocorticosteroids, eculizumab, rituximab, maintenance intravenous immunoglobulin (IVIG) or plasma exchange, and cyclophosphamide); 3) rapid but transient immunomodulatory therapies (plasma exchange and intravenous immune globulin) and 4) thymectomy. Approximately 10 percent of individuals with gMG have symptoms that are refractory or limited by specific toxicities of conventional immunomodulatory therapies (eg, high-dose glucocorticoids). Therapeutic options for refractory disease include azathioprine, cyclosporine, eculizumab, efgartigimod, mycophenolate, ravulizumab, and tacrolimus.

In order to stabilize a patient with myasthenia gravis an operative procedure, IVIG or plasmapheresis may be utilized. These interventions are also the treatment of choice during a myasthenic crisis and in individuals who are resistant to immunosuppressive medications. Thymectomy may be employed as a therapeutic approach for certain individuals with myasthenia gravis.

Regulatory Status

On October 23, 2017, eculizumab (Soliris®, Alexion Pharmaceuticals Inc) was approved by the U.S. FDA for the treatment of gMG in adult patients who are anti-acetylcholine receptor antibody positive.

On December 17, 2021, efgartigimod alfa-fcab (Vyvgart®, Argenx BV) was approved by the U.S. FDA for the treatment of gMG in adult patients who are anti-acetylcholine receptor antibody positive. On June 20, 2023, U.S. FDA approved a subcutaneous formulation (Vyvgart Hytrulo, Argenx BV) for the same indication.

On April 28, 2022, ravulizumab-cwvz (Ultomiris®, Alexion Pharmaceuticals Inc) was approved by the U.S. FDA for the treatment of gMG in adult patients who are anti-acetylcholine receptor antibody positive.

On June 26, 2023, rozanolixizumab-noli (Rystiggo®, UCB Inc) was approved by the U.S. FDA for the treatment of gMG in adult patients who are anti-acetylcholine receptor or antimuscle-specific tyrosine kinase antibody positive.

RATIONALE

This evidence review was created in May 2022 with a search of the PubMed database. The most recent literature update was performed through April 9, 2024.

Evidence reviews assess the clinical evidence to determine whether the use of a technology improves the net health outcome. Broadly defined, health outcomes are 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 to 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 a technology, 2 domains are examined: the relevance and the 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, nonrandomized studies may be adequate. Randomized controlled trials 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.

Generalized Myasthenia Gravis

Clinical Context and Therapy Purpose

The purpose of eculizumab in adults who have gMG who are anti-acetylcholine receptor antibody positive is to provide a treatment option that is an alternative to existing therapeutic management for individuals with refractory disease.

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

Populations

The relevant population(s) of interest is adults with gMG who are anti-acetylcholine receptor antibody positive.

Interventions

The therapies being considered are eculizumab, ravulizumab-cwvz, efgartigimod, and rozanolixizumab.

Eculizumab and ravulizumab-cwvz are monoclonal antibodies that are presumed to exert a therapeutic effect in individuals with gMG through the reduction of terminal complement complex C5b-9 deposition at the neuromuscular junction. Rozanolixizumab-noli is also a humanized IgG4 monoclonal antibody that binds to the neonatal Fc receptor resulting in the reduction of circulating IgG. Efgartigimod alfa-fcab is a human IgG1 antibody fragment that binds to the neonatal Fc receptor, resulting in the reduction of circulating IgG in individuals with gMG.

Comparators

The following therapies are currently being used to make decisions about the treatment of gMG: acetylcholinesterase inhibitors, immunosuppressive agents, monoclonal antibodies, intravenous immunoglobulin/plasmapheresis, and thymectomy. Treatment is dependent upon response to therapy, setting (eg, preoperative), presence of myasthenic crisis, and etiology of myasthenia gravis.

Outcomes

The general outcomes of interest are symptoms, quality of life, hospitalizations, and resource utilization. Health outcome measures relevant to gMG in adults are summarized in Table 1.

Follow-up of months to years is of interest to monitor outcomes.

Table 1. Health Outcome Measures Relevant to Generalized Myasthenia Gravis in Adults10,
Outcome Description and Administration Thresholds for Improvement/Decline or Clinically Meaningful Difference (if known)
Quantitative Myasthenia Gravis
  • Measure of disease severity; includes 13 items that assess muscle strength and fatigability using objective measures of double vision, ptosis, facial muscles, dysphagia, dysarthria, proximal limb, hand muscles, neck muscles, and respiratory function
  • Assessments are time consuming and require equipment; use in clinical practice is challenging
  • Each item is given a score of 0-3, resulting in an unweighted total score of 0-39; a higher score corresponds to more severe disease
  • A 3-point change is considered clinically meaningful, with a modification in milder cases where a 2-point change is considered sufficient
Myasthenia Gravis-Activity of Daily Living
  • Patient-reported outcome; quickly administered set of questions examining frequency and severity of key myasthenia gravis symptoms
  • Eight questions assessing ocular function, speech, chewing, swallowing, respiratory function, and strength of proximal upper and lower extremities
  • Each item is scored from 0-3, which results in an unweighted total score of 0-24 points; a higher score indicates more severe symptoms
  • A 2-point change is considered clinically meaningful
Myasthenia Gravis Composite
  • Uses the top performing items of the Quantitative Myasthenia Gravis, Myasthenia Gravis-Activity of Daily Living, and the Manual Muscle Test; 6 physician-assessed examinations evaluate ocular, neck, and proximal limb muscles with 4 patient-reported items assessing speech, chewing, swallowing, and respiratory function
  • Total score spans from 0-50; a higher score indicating more severe disease
  • A 3-point change is considered clinically meaningful based on physician's impression of change
Myasthenia Gravis Quality of Life 15-items
  • Patient reported 15 questions assess ocular symptoms, swallowing, speech, proximal limb function, mobility, personal grooming, work, social life, activities, fluctuations, and psychological items
  • Each question is scored from 0-4, resulting in a total score in the range of 0-60; a higher score indicates poorer quality of life
  • QOL15 was slightly revised to the current QOL15r that retains the original questions and reduces the item score to a range of 0-2
Abbreviations: QOL: quality of life.
 
Study Selection Criteria

Methodologically credible studies were selected using the following principles:

Review of Evidence

Population Reference No. 1

Eculizumab

Randomized Controlled Trials

Trial characteristics and results of the pivotal double-blind, placebo-controlled, phase 3 REGAIN trial are summarized in Tables 2 and 3, respectively.11, The study met the primary efficacy endpoint. A statistically significant difference favoring eculizumab was observed in the mean change from baseline to week 26 in Myasthenia Gravis-Specific Activities of Daily Living scale (MG-ADL) total scores [-4.2 points in the eculizumab-treated group compared with -2.3 points in the placebo-treated group (p=.006)]. A key secondary endpoint of change from baseline in the Quantitative Myasthenia Gravis (QMG) total score at week 26 also favored eculizumab [-4.6 points in the eculizumab-treated group compared with -1.6 points in the placebo-treated group (p=.001)].12, An open-label extension of the pivotal REGAIN trial reported data from a preplanned interim analysis that was based on a median duration of approximately 2 years of eculizumab therapy (N=117).13, Results showed that individuals who had received placebo during the REGAIN double-blind phase experienced rapid and sustained improvements during the open-label eculizumab phase. Compared with the year before the REGAIN trial started (pre-study baseline), the myasthenia gravis exacerbation rate was reduced by 75.2% (pre-study, 102.4 exacerbations per 100 patient‐years; open‐label study, 25.4 exacerbations per 100 patient‐years; p<.0001). Long-term follow-up based on 227 patient‐years of open‐label eculizumab exposure reported that the safety and efficacy of eculizumab was sustained with long‐term treatment.13,

Eculizumab was issued a boxed warning due to the life-threatening and fatal meningococcal infections that occurred in individuals treated with eculizumab. These infections may become rapidly life-threatening or fatal if not recognized and treated early. The most frequently reported adverse reaction (≥10%) is musculoskeletal pain.12,

Table 2. Summary of Pivotal RCT Characteristics
Study Countries Sites Dates Participants Interventions
          Active Comparator
REGAIN12,(NCT01997229) 17 across North America, Latin America, Europe, and Asia 76 April 2014-February 2016 Inclusion
  • Positive serologic test for anti-AChR antibodies
  • MGFA clinical classification class II to IV
  • MG-ADL total score ≥6
  • Failed treatment over 1 year or more with 2 or more immunosuppressive therapies either in combination or as monotherapy, or failed at least 1 immunosuppressive therapy and required chronic plasmapheresis or plasma exchange or intravenous immunoglobulin
Primary Endpoint
  • Change from baseline between treatment groups in the MG-ADL total score at week 26
Eculizumab 900 mg IV on day 1 and weeks 1, 2, and 3; 1200 mg at week 4; and maintenance dosing of 1200 mg every other week thereafter for 26 weeks (n=62) Placebo given on the same schedule (n=63)
AChR: acetylcholine receptor; gMG: generalized myasthenia gravis; IV: intravenous; IVIG: intravenous immunoglobulin; MGFA: Myasthenia Gravis Foundation of America; MG-ADL: Myasthenia Gravis-Activities of Daily Living; RCT: randomized controlled trial.
 
Table 3. Summary of Pivotal RCT Results
Study Eculizumab Placebo
REGAIN12,14,    
N 62 63
Primary endpoint    
LS mean change from baseline to week 26 in MG-ADL total scores, mean (±SEM) -4.2 (0.49) -2.3 (0.48)
LS mean difference in eculizumab relative to placebo (95% CI) -1.9 (-3.3 to -0.6)
p-value .006a,.014b
Secondary endpoints    
LS change from baseline to week 26 in QMG total scores, mean (±SEM) -4.6 (0.60) -1.6 (0.59)
LS mean difference in eculizumab relative to placebo (95% CI) -3.0 (-4.6 to -1.3)
p-value .001a,.005b
Responder analysis    
≥3-point reduction in MG‐ADL total score from baseline to week 26, n (%) 37 (60%) 25 (40%)
p-value .02
≥5-point reduction in QMG total score from baseline to week 26, n (%) 28 (45%) 12 (19%)
p-value .002
CI: confidence interval; LS: least square; MG-ADL; Myasthenia Gravis-Activity of Daily Living; QMG: Quantitative Myasthenia Gravis; RCT: randomized controlled trial. SEM: standard error of mean.
a p-values testing the null hypothesis that there is no difference in least square means at week 26 using a repeated measure analysis
b p-values testing the null hypothesis that there is no difference in ranks at week 26 using a worst rank analysis

The purpose of the study limitations table (Table 4) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence following the table and provides the conclusions on the sufficiency of evidence supporting the position statement. The limited representations of African Americans, Asians, and Hispanics makes it challenging to reach conclusions about the efficacy of eculizumab in these racial groups. No major limitations in the study design and conduct were identified.

Table 4. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Duration of Follow-upe
REGAIN 4. 76% White and 15% Asian        
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. Study population is unclear; 3. Study population not representative of intended use; 4, Enrolled populations do not reflect relevant diversity; 5. Other.
b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest (e.g., proposed as an adjunct but not tested as such); 5: Other.
c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively; 5. Other.
d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. Incomplete reporting of harms; 4. Not establish and validated measurements; 5. Clinically significant difference not prespecified; 6. Clinically significant difference not supported; 7. Other.
e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms; 3. Other.

Sub-section Summary: Eculizumab

Results of the pivotal RCT REGAIN reported a statistically significant difference in the primary endpoint favoring eculizumab in MG-ADL total scores compared with the placebo (least square (LS) mean difference of -1.9 points; 95% CI: -3.3 to -0.6). A key secondary endpoint of change from baseline in the QMG total score at week 26 also favored eculizumab compared with placebo (LS mean difference of -3.0 points; 95% CI: -4.6 to -1.3). The proportion of responders as defined by at least a 3-point reduction in MG‐ADL total score and at least a 5-point reduction in QMG total score from baseline to week 26 in eculizumab compared to placebo was 60% versus 40% and 45% versus 19%, respectively. An open-label extension of the pivotal trial provided additional evidence that showed that individuals who had received placebo during the REGAIN double-blind phase experienced rapid and sustained improvements during the open-label eculizumab phase. Eculizumab was issued a boxed warning due to the life-threatening and fatal meningococcal infections that occurred in individuals treated with eculizumab and may become rapidly life-threatening or fatal if not recognized and treated early. The most frequently reported adverse reaction (≥10%) is musculoskeletal pain. The limited representations of African Americans, Asians, and Hispanics makes it challenging to reach conclusions about the efficacy of eculizumab in these racial groups. No major limitations in the study design and conduct were identified.

For individuals with generalized myasthenia gravis who receive eculizumab, the evidence includes a single pivotal randomized controlled trial (RCT). Relevant outcomes are symptoms, quality of life, hospitalizations, and resource utilization. Results of the pivotal REGAIN trial reported a statistically significant difference in the primary endpoint favoring eculizumab in Myasthenia Gravis-Specific Activities of Daily Living scale (MG-ADL) total scores compared with the placebo (least square mean difference of -1.9 points; 95% confidence interval [CI]: -3.3 to -0.6). A key secondary endpoint of change from baseline in the Quantitative Myasthenia Gravis (QMG) total score at week 26 also favored eculizumab compared with placebo (least square mean difference of -3.0 points; 95% CI: -4.6 to -1.3). Proportion of responders as defined by at least a 3-point reduction in MG‐ADL total score and at least a 5-point reduction in QMG total score from baseline to week 26 in eculizumab compared to placebo was 60% versus 40% and 45% versus 19%, respectively. An open-label extension of the pivotal trial provided additional evidence that showed that individuals who had received placebo during the REGAIN double-blind phase experienced rapid and sustained improvements during open-label eculizumab phase. 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] Medically Necessary

[ ] Investigational

Population Reference No. 2

Ravulizumab

Randomized Controlled Trials

Trial characteristics and results of the pivotal double-blind, placebo-controlled, phase 3 CHAMPION MG trial are summarized in Tables 5 and 6, respectively. 15,The study met the primary efficacy endpoint. A statistically significant difference favoring ravulizumab was observed in the mean change from baseline to week 26 in MG-ADL total scores [-3.1 points in the ravulizumab-treated group compared with -1.4 points in the placebo-treated group (p=.001)]. A key secondary endpoint of change from baseline in the QMG total score at week 26 also favored ravulizumab [-2.8 points in the ravulizumab-treated group compared with -0.8 points in the placebo-treated group (p=.001)].16, Responder analysis also favored the ravulizumab treated arm compared to placebo. Results from the open-label extension phase reported that improvements in all scores were maintained through 60 weeks; LS mean change from baseline in MG-ADL score was -4.0 (95% CI: -4.8 to - 3.1; p<.0001).17,

Ravulizumab was issued a boxed warning due to the life-threatening and fatal meningococcal infections that occurred in individuals treated with eculizumab and may become rapidly life-threatening or fatal if not recognized and treated early. The most frequently reported adverse reactions (≥10%) were upper respiratory tract infection and headache.16,

Table 5. Summary of Pivotal RCT Characteristics
Study Countries Sites Dates Participants Interventions
          Active Comparator
CHAMPION MG16, NCT03920293 Global 85 March 2019-May 2021
Inclusion
  • Positive serologic test for anti-AChR antibodies
  • MGFA clinical classification class II to IV
  • MG-ADL total score ≥6
Primary Endpoint
  • Change from baseline between treatment groups in the MG-ADL total score at week 26
Ravulizumab IV weight-based dosing; initial loading dose of 2400, 2700, or 3000 mg at day 1, followed by maintenance doses of 3000, 3300, or 3600 mg on day 15 and every 8 weeks thereafter for 26 weeks (n=86) Placebo given on the same schedule (n=89)
AChR: acetylcholine receptor; IV: intravenous; MG-ADL: Myasthenia Gravis-Activity of Daily Living; MGFA: Myasthenia Gravis Foundation of America; RCT: randomized controlled trial.
 
Table 6. Summary of Pivotal RCT Results
Study Ravulizumab Placebo
CHAMPION MG16,    
N 86 63
Primary endpoint    
LS mean change from baseline to week 26 in MG-ADL total scores -3.1 -1.4
LS mean difference in eculizumab relative to placebo (95% CI) -1.6 (-2.6 to -0.7)
p-value .001
Secondary endpoints    
LS change from baseline to week 26 in QMG total scores -2.8 -0.8
LS mean difference in eculizumab relative to placebo (95% CI) -2.0 (-3.2 to -0.8)
p-value .001
Responder analysis    
≥3-point reduction in MG‐ADL total score from baseline to week 26 57% 34%
p-value .005
≥5-point reduction in QMG total score from baseline to week 26 30% 11%
p-value .005
CI: confidence interval; LS: least squares; MG-ADL: Myasthenia Gravis-Activity of Daily Living; QMG: Quantitative Myasthenia Gravis; RCT: randomized controlled trial.
p-value calculated using mixed effect model for repeated measures

The purpose of the study limitations table (Table 7) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence following the table and provides the conclusions on the sufficiency of evidence supporting the position statement. The limited representations of African Americans, Asians, and Hispanics makes it challenging to reach conclusions about the efficacy of ravulizumab in these racial groups. No major limitations in the study design and conduct were identified.

Table 7. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Duration of Follow-upe
CHAMPION MG16, 4. Enrolled populations do not reflect relevant diversity (73% White and 18% Asian)      
 
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. Study population is unclear; 3. Study population not representative of intended use; 4, Enrolled populations do not reflect relevant diversity; 5. Other.
b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest (e.g., proposed as an adjunct but not tested as such); 5: Other.
c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively; 5. Other.
d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. Incomplete reporting of harms; 4. Not establish and validated measurements; 5. Clinically significant difference not prespecified; 6. Clinically significant difference not supported; 7. Other.
e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms; 3. Other.

Sub-section Summary: Ravulizumab-cwvz

Results of the pivotal RCT CHAMPION MG reported a statistically significant difference in the primary endpoint favoring ravulizumab in MG-ADL total scores compared with the placebo (LS mean difference of -1.6 points; 95% CI: -2.6 to -0.7). A key secondary endpoint of change from baseline in the QMG total score at week 26 also favored ravulizumab compared with placebo (LS mean difference of -2.0 points; 95% CI: -3.2 to -0.8). The proportion of responders as defined by at least a 3-point reduction in MG‐ADL total score and at least a 5-point reduction in QMG total score from baseline to week 26 in eculizumab compared to placebo was 57% versus 34% and 30% versus 11%, respectively. Ravulizumab was issued a boxed warning due to the life-threatening and fatal meningococcal infections that occurred in individuals treated with ravulizumab and may become rapidly life-threatening or fatal if not recognized and treated early. The most frequently reported adverse reactions (≥10%) were upper respiratory tract infection and headache. The limited representations of African Americans, Asians, and Hispanics makes it challenging to reach conclusions about the efficacy of ravulizumab in these racial groups. No major limitations in the study design and conduct were identified.

For individuals with generalized myasthenia gravis who receive ravulizumab, the evidence includes a single pivotal RCT. Relevant outcomes are symptoms, quality of life, hospitalizations, and resource utilization. Results of the pivotal CHAMPION MG trial reported a statistically significant difference in the primary endpoint favoring ravulizumab in MG-ADL total scores compared with the placebo (least square mean difference of -1.6 points; 95% CI: -2.6 to -0.7). A key secondary endpoint of change from baseline in the QMG total score at week 26 also favored ravulizumab compared with placebo (least square mean difference of -2.0 points; 95% CI: -3.2 to -0.8). Proportion of responders as defined by at least a 3-point reduction in MG‐ADL total score and at least a 5-point reduction in QMG total score from baseline to week 26 in eculizumab compared to placebo was 57% versus 34% and 30% versus 11%, respectively. 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] Medically Necessary

[ ] Investigational

Population Reference No. 3

Efgartigimod

Two formulations are currently approved by the FDA – Vygart (efgartigimod alfa-fcab injection for intravenous infusion) and Vygart Hytrulo (efgartigimod alfa and hyaluronidase-qvfc for subcutaneous use). The US FDA approval of the intravenous formulation was based on a 26-week, double-blind, placebo-controlled randomized trial called ADAPT, while the approval for the subcutaneous formulation was based on the results of a bridging 10-week, open-label, randomized trial called ADAPT-SC. These are summarized next.

Randomized Controlled Trials

Trial characteristics and results of the pivotal trials are summarized in Tables 8 and 9, respectively. 18,19, ADAPT was a double-blind, placebo-controlled, phase 3 trial. While it enrolled individuals regardless of anti-AChR antibody status, the primary endpoint and subsequent approval of efgartigimod by the FDA was only for individuals who were AChR-Ab positive. The study met the primary efficacy endpoint. A statistically significant difference favoring efgartigimod was observed in the MG-ADL responder rate [67.7% in the efgartigimod -treated group vs 29.7% in the placebo-treated group (p<.0001)]. A key secondary endpoint of comparison of the proportion of QMG responders between the 2 treatment groups also favored efgartigimod [63.1% in the efgartigimod-treated group vs 14.1% in the placebo-treated group (p<.0001)]. The most frequently reported adverse reactions (≥10%) were respiratory tract infections, headache, and urinary tract infection.

ADAPT-SC was a randomized open-label parallel-group trial with the objective to demonstrate pharmacodynamic non-inferiority of the subcutaneous formulation to that of the intravenous formulation. It also enrolled individuals regardless of anti-AChR antibody status. The noninferiority evaluation was based on the percent reduction from baseline in AChR-Ab levels at day 29 (ie, week 4) using an noninferiority margin of 10% meaning that when the lower limit of the 95% confidence interval for the difference is above the margin of -10, the subcutaneous formulation will be considered noninferior to the IV formulation.20, The LS mean difference in the percent change from baseline of AChR-Ab levels was 2.5% (95% CI: -7.45 to 2.41), which is below the upper limit of the confidence interval of 10%.21, Additionally, the 90% CIs for the geometric mean ratios of AChR-Ab reduction at day 29 and AUEC0-4w (area under the effect-time curve from time 0 to 4 weeks post dose) were within the range of 80% to 125%, indicating no clinically significant difference between the two formulations.22,

Table 8. Summary of Pivotal RCT Characteristics
Study Countries Sites Dates Participants Interventions
          Active Comparator
ADAPT18, (NCT03669588) Global 56 September 2018-November 2019
Inclusion
  • MGFA clinical classification class II to IV
  • MG-ADL total score ≥5
  • On stable dose of myasthenia gravis treatment prior to screening, that included acetylcholinesterase inhibitors, steroids, or NSISTs, either in combination or alone
  • IgG levels ≥6 g/L
Primary endpoint
  • Change in proportion of MG-ADL responders from baseline between treatment groups at week 26 in the AChR-Ab positive populationa
Efgartigimod 10 mg/kg IV administered as 4 infusions per cycle (1 infusion per week) repeated as needed depending on clinical response no sooner than 8 weeks after initiation of the previous cycle for 26 weeks (n=84) Placebo given on the same schedule (n=83)
ADAPT-SC22, (NCT04735432) Global 47 Feb 2021-Dec 2021 Inclusion
  • MGFA clinical classification class II to IV
  • MG-ADL total score of ≥5 with >50% of the total score attributed to nonocular symptoms.
  • All individuals received stable doses of their current gMG treatment.
Primary endpoint
  • Percent reduction from baseline in total immunoglobulin G levels at day 29 (that is 7 days after the fourth IV or SC administration)
Efgartigimod alfa 1008 mg/11,200 units of hyaluronidase subcutaneous injection once weekly for 4 week (n=55) Efgartigimod alfa-fcab 10 mg/kg IV administered once weekly for 4 weeks (n=55)
AChR: acetylcholine receptor; gMG: generalized myasthenia gravis; IV: intravenous; MG-ADL: Myasthenia Gravis-Activity of Daily Living; MGFA: Myasthenia Gravis Foundation of America; NSISTs: non-steroidal immunosuppressive therapies; RCT: randomized controlled trial; SC: subcutaneous.
a MGADL responder was defined as a patient with a 2-point or greater reduction in the total MG-ADL score compared to the treatment cycle baseline for at least 4 consecutive weeks, with the first reduction occurring no later than 1 week after the last infusion of the cycle
Table 9. Summary of Pivotal RCT Results
Study Efgartigimod Placebo
ADAPT18,    
N 65 67
Primary endpoint    
MG‐ADL responders among AChR-Ab positive patientsa 67.7 29.7
p-value .0001
OR (95% CI) 4.95 (2.21 to 11.53)
Secondary endpoints    
QMG responders among AChR-Ab positive individualsb 63.1 14.1
p-value .0001
OR (95% CI) 10.84 (4.18 to 31.20)
ADAPT-SC22,21, Efgartigimod alfa plus hyaluronidase Efgartigimod alfa
N 44 42
Percent reduction from baseline in AChR-Ab levels at week 4 (day 29) among AChR-Ab positive individuals 62.2% 59.7%
LSM difference 2.5% (95% CI: -7.45 to 2.41),
Ab: antibody; AChR: acetylcholine receptor; CI: confidence interval; MG-ADL: Myasthenia Gravis-Activity of Daily Living; OR: odds ratio; QMG: Quantitative Myasthenia Gravis; RCT: randomized controlled trial.
CI: confidence interval; LSM: least squares mean; MG-ADL: Myasthenia Gravis-Activity of Daily Living; QMG: Quantitative Myasthenia Gravis; RCT: randomized controlled trial.
p-value calculated using mixed effect model for repeated measures
a MG-ADL responder was defined as a patient with a 2-point or greater reduction in the total MG-ADL score compared to the treatment cycle baseline for at least 4 consecutive weeks, with the first reduction occurring no later than 1 week after the last infusion of the cycle
b QMG responder was defined as a patient who had a 3-point or greater reduction in the total QMG score compared to the treatment cycle baseline for at least 4 consecutive weeks, with the first reduction occurring no later than 1 week after last infusion of the cycle.

The purpose of the study limitations table (Table 10) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence following the table and provides the conclusions on the sufficiency of evidence supporting the position statement. The limited representations of African Americans, Asians, and Hispanics makes it challenging to reach conclusions about the efficacy of efgartigimod in these racial groups. Because of the relatively short follow-up, there is still considerable uncertainty about the long-term net benefits of efgartigimod compared with other treatment options. No major limitations in the study design and conduct were identified.

Table 10. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Duration of Follow-upe
ADAPT18, 4. Enrolled populations do not reflect relevant diversity (88% White)
 
      1. Not sufficient duration for benefit;
2. Not sufficient duration for harms;
(study duration limited to 26 weeks)
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. Study population is unclear; 3. Study population not representative of intended use; 4, Enrolled populations do not reflect relevant diversity; 5. Other.
b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest (e.g., proposed as an adjunct but not tested as such); 5: Other.
c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively; 5. Other.
d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. Incomplete reporting of harms; 4. Not establish and validated measurements; 5. Clinically significant difference not prespecified; 6. Clinically significant difference not supported; 7. Other.
e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms; 3. Other.

Sub-section Summary: Efgartigimod

Two formulations are currently approved by the FDA: Vygart (efgartigimod alfa-fcab injection for intravenous infusion) and Vygart Hytrulo (efgartigimod alfa and hyaluronidase-qvfc for subcutaneous use). Initial FDA approval of the intravenous formulation was based on a single RCT called ADAPT. Results of this trial reported a statistically significant difference in the primary endpoint favoring efgartigimod in MG-ADL responder rate compared with the placebo (67.7% vs 29.7%, respectively; p<.0001). A key secondary endpoint of responder based on QMG total score at week 26 also favored efgartigimod compared with placebo (63.1% vs 14.1%, respectively; p<.0001). The most frequently reported adverse reactions (≥10%) were respiratory tract infections, headache, and urinary tract infection. Subsequent approval of the subcutaneous formulation was based on the results of a bridging 10-week open-label randomized trial called ADAPT-SC. Results of this trial demonstrated pharmacodynamic non-inferiority based on the percent reduction in AChR-Ab levels from baseline to day 29. The LS mean difference was 2.5% (95% CI: -7.45 to 2.41), which was below the upper limit of the confidence interval of 10%. The limited representations of African Americans, Asians, and Hispanics in the ADAPT trial make it challenging to reach conclusions about the efficacy of efgartigimod in these racial groups. Because of the relatively short follow-up, there is still considerable uncertainty about the long-term net benefits of efgartigimod compared with other treatment options. No major limitations in the study design and conduct were identified.

For individuals with generalized myasthenia gravis who receive efgartigimod, the evidence includes 2 pivotal RCTs. Relevant outcomes are symptoms, quality of life, hospitalizations, and resource utilization. Two formulations are currently approved by the Food and Drug Administration (FDA): Vygart (efgartigimod alfa-fcab injection for intravenous infusion) and Vygart Hytrulo (efgartigimod alfa and hyaluronidase-qvfc for subcutaneous use). Initial FDA approval of the intravenous formulation was based on a single RCT called ADAPT. Results of this trial reported a statistically significant difference in the primary endpoint favoring efgartigimod in MG-ADL responder rate compared with the placebo (67.7% versus 29.7%, respectively; p<.0001). MG-ADL responder was defined as a patient with a 2-point or greater reduction in the total MG-ADL score compared to the treatment cycle baseline for at least 4 consecutive weeks, with the first reduction occurring no later than 1 week after the last infusion of the cycle. A key secondary endpoint of responder based on QMG total score at week 26 also favored efgartigimod compared with placebo (63.1% versus 14.1%, respectively; p<.0001). QMG responder was defined as a patient who had a 3-point or greater reduction in the total QMG score compared to the treatment cycle baseline for at least 4 consecutive weeks, with the first reduction occurring no later than 1 week after the last infusion of the cycle. Subsequent approval of the subcutaneous formulation was based on the results of a bridging 10-week open-label randomized trial called ADAPT-SC. Results of this trial demonstrated pharmacodynamic non-inferiority based on the percent reduction in AChR-Ab levels from baseline to day 29. The least squares mean difference was 2.5% (95% CI: -7.45 to 2.41), which was below the upper limit of the confidence interval of 10%. Because of the relatively short follow-up, there is still considerable uncertainty about the long-term net benefits of efgartigimod compared with other treatment options. No major limitations in the study design and conduct were identified. 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] Medically Necessary

[ ] Investigational

Population Reference No. 4

Rozanolixizumab

Randomized Controlled Trials

Trial characteristics and results of the pivotal double-blind, placebo-controlled, phase 3 MycarinG trial are summarized in Tables 11 and 12, respectively. The study met the primary efficacy endpoint. A statistically significant difference favoring rozanolixizumab was observed in the mean change from baseline to day 43 in MG-ADL total scores [-3.4 points in the rozanolixizumab-treated group either doses compared with -0.8 points in the placebo-treated group (p<.001)]. A key secondary endpoint of change from baseline in the QMG total score at week 43 also favored rozanolixizumab [-5.4 points and -6.7 points in the rozanolixizumab-treated group at 7 mg/kg and 10 mg/kg dose level, respectively, compared with -1.9 points in the placebo-treated group (p<.001)]. The most common adverse reactions (≥10%) were headache, infections, diarrhea, pyrexia, hypersensitivity reactions, and nausea. Serious events of aseptic meningitis were reported. As per the label, symptoms for meningitis should be monitored and diagnostic workup and treatment should be initiated according to the standard of care.23,

Table 11. Summary of Pivotal RCT Characteristics
Study Countries Sites Dates Participants Interventions
          Active Comparator
MycarinG23,24,(NCT03971422) Global 112 June 2019-June 2021
Inclusion
  • Presence of autoantibodies against AChR or MuSK
  • MGFA clinical classification class II to IVa
  • MG-ADL total score ≥3 (with ≥3 points from non-ocular symptoms)
  • On stable dose of myasthenia gravis therapy prior to screening that included acetylcholinesterase inhibitors, steroids, or non-steroidal immunosuppressive therapies, either in combination or alone
  • Serum IgG levels of ≥5.5 g/L
Primary Endpoint
  • Change from baseline in the MG-ADL total score from baseline to day 43
Rozanolixizumab subcutaneously once a week for 6 weeks (n=133); 7 mg/kg (n=66), or 10 mg/kg (n=67) Placebo given on the same schedule (n=67)
AChR: acetylcholine receptor; IV: intravenous; MG-ADL: Myasthenia Gravis-Activity of Daily Living; MGFA: Myasthenia Gravis Foundation of America; MuSK: antimuscle-specific tyrosine kinase; RCT: randomized controlled trial.
 
Table 12. Summary of Pivotal RCT Results
Study Rozanolixizumab 7 mg/kg Rozanolixizumab 10 mg/kg Placebo
MycarinG23,24,      
N 66 67 67
Primary endpoint (MG-ADL Total Score)      
LS mean change from baseline to day 41 (SE) -3.4 (0.5) -3.4 (0.5) -0.8 (0.5)
Difference from placebo (95% CI) -2.6 (-4.1 to -1.2) -2.6 (-4.0 to -1.2) -
p-value .001 .001 -
Secondary endpoint (QMG Total Score)      
LS mean change from baseline to day 41 (SE) -5.4 (0.7) -6.7 (0.7) -1.9 (0.7)
Difference from placebo (95% CI) -3.5 (-5.6 to -1.6) -4.8 (-6.8 to -2.9) -
p-value .001 .001 -
CI: confidence interval; LSM: least squares mean; MG-ADL: Myasthenia Gravis-Activity of Daily Living; QMG: Quantitative Myasthenia Gravis; RCT: randomized controlled trial; SE: standard error.

The purpose of the study limitations table (Table 13) is to display notable limitations identified in each study. This information is synthesized as a summary of the body of evidence following the table and provides the conclusions on the sufficiency of evidence supporting the position statement. The limited representations of African Americans, Asians, and Hispanics and anti-MuSK antibody positive individuals makes it challenging to reach conclusions about the efficacy of rozanolixizumab in these groups. Because of the relatively short follow-up, there is still considerable uncertainty about the long-term net benefits of rozanolixizumab compared with other treatment options. No major limitations in the study design and conduct were identified.

Table 13. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Duration of Follow-upe
MycarinG23,24, 3. Study population not representative of intended use (<10% of trial participants were anti-MuSK positive)
4. Enrolled populations do not reflect relevant diversity (68% White, 11% Asian, 7% Hispanic or Latino, 3% Black or African American)
      1. Not sufficient duration for benefit;
2. Not sufficient duration for harms;
(study duration limited to 8 weeks)
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. Study population is unclear; 3. Study population not representative of intended use; 4, Enrolled populations do not reflect relevant diversity; 5. Other.
b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4. Not the intervention of interest (e.g., proposed as an adjunct but not tested as such); 5: Other.
c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively; 5. Other.
d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. Incomplete reporting of harms; 4. Not establish and validated measurements; 5. Clinically significant difference not prespecified; 6. Clinically significant difference not supported; 7. Other.
e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms; 3. Other.

Sub-section Summary: Rozanolixizumab

Results of the pivotal RCT MycarinG reported a statistically significant difference in the primary endpoint favoring rozanolixizumab in MG-ADL total scores compared with the placebo (LS mean difference of -2.6 points for either doses of rozanolixizumab). A key secondary endpoint of change from baseline in the QMG total score at week 43 also favored rozanolixizumab [-5.4 points and -6.7 points in the rozanolixizumab-treated group at 7 mg/kg and 10 mg/kg dose level, respectively compared with -1.9 points in the placebo-treated group (p<.001)]. The most common adverse reactions (≥10%) were headache, infections, diarrhea, pyrexia, hypersensitivity reactions, and nausea. Serious events of aseptic meningitis were reported. The limited representations of African Americans, Asians, and Hispanics makes it challenging to reach conclusions about the efficacy of rozanolixizumab in these racial groups. Because of relatively short follow-up, there is still considerable uncertainty about the long-term net benefits of rozanolixizumab compared with other treatment options. No major limitations in the study design and conduct were identified.

For individuals with generalized myasthenia gravis who receive rozanolixizumab, the evidence includes a single pivotal RCT. Relevant outcomes are symptoms, quality of life, hospitalizations, and resource utilization. Results of the pivotal RCT MycarinG reported a statistically significant difference in the primary endpoint favoring rozanolixizumab in MG-ADL total scores compared with the placebo (LS mean difference of -2.6 points for either dose of rozanolixizumab). A key secondary endpoint of change from baseline in the QMG total score at week 43 also favored rozanolixizumab [-5.4 points and -6.7 points in the rozanolixizumab-treated group at 7 mg/kg and 10 mg/kg dose level, respectively, compared with -1.9 points in the placebo-treated group (p<.001)]. Because of the relatively short follow-up, there is still considerable uncertainty about the long-term net benefits of rozanolixizumab compared with other treatment options. 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] Medically Necessary

[ ] 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.

Myasthenia Gravis Foundation of America

In 2021, an international consensus guidance for the management of myasthenia gravis was published.25, This guidance contained recommendations for the use of eculizumab including:

U.S. Preventive Services Task Force Recommendations

No U.S. Preventive Services Task Force (USPSTF) recommendations for the treatment of generalized myasthenia gravis have been identified.

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 trials that might influence this review are listed in Table 14.

Table 14. Summary of Key Trials
NCT No. Trial Name Planned Enrollment Completion Date
Ongoing      
Eculizumab      
NCT04202341a Registry of Participants With Generalized Myasthenia Gravis Treated With Alexion C5 Inhibition Therapies (C5ITs) 500 Dec 2029
Efgartigimod      
NCT04833894a Open-Label Uncontrolled Trial to Evaluate Pharmacokinetics, Pharmacodynamics, Safety, and Activity of Efgartigimod in Children From 2 to Less Than 18 Years of Age With Generalized Myasthenia Gravis 12 Aug 2024
NCT04818671a A Long-Term, Single-Arm, Open-Label, Multicenter Phase 3 Study to Evaluate the Safety and Tolerability of Multiple Subcutaneous Injections of Efgartigimod PH20 SC in Patients With Generalized Myasthenia Gravis 183 Dec 2024
NCT04980495a A Phase 3b, Randomized, Open-Label, Parallel-Group Study to Evaluate Different Dosing Regimens of Intravenous Efgartigimod to Maximize and Maintain Clinical Benefit in Patients With Generalized Myasthenia Gravis 69 May 2026
NCT05374590 A Long-term, Single-Arm, Open-label, Multicenter, Follow-on Trial of ARGX-113-2006 to Evaluate Safety of Efgartigimod Administered Intravenously in Children With Generalized Myasthenia Gravis 12 Sep 2028
Ravulizumab      
NCT05644561 A Phase 3, Open-label, Single-arm, Multicenter Study to Evaluate the Pharmacokinetics, Pharmacodynamics, Efficacy, Safety, and Immunogenicity of Ravulizumab Administered Intravenously in Pediatric Participants (6 to < 18 Years of Age) With Generalized Myasthenia Gravis (gMG) 12 Jul 2028
Rozanolixizumab      
NCT05681715 An Open-label, Crossover Study to Evaluate Rozanolixizumab Self-administration by Study Participants With Generalized Myasthenia Gravis 62 Apr 2024
NCT04650854 An Open-Label Extension Study to Evaluate Rozanolixizumab in Study Participants With Generalized Myasthenia Gravis 165 Jan 2024
Unpublished      
NCT04735432 (ADAPTsc) Evaluating the Pharmacodynamic Noninferiority of Efgartigimod PH20 SC Administered Subcutaneously as Compared to Efgartigimod Administered Intravenously in Patients With Generalized Myasthenia Gravis (ADAPTsc) 110 Dec 2021
NCT04124965 A Study to Investigate the Long-term Safety, Tolerability, and Efficacy of Rozanolixizumab in Adult Patients With Generalized Myasthenia Gravis 71 Sep 2021
NCT03052751 Study to Test the Safety, Tolerability and Efficacy of UCB7665 in Subjects With Moderate to Severe Myasthenia Gravis 43 Aug 2018
NCT03759366a A Phase 3 Open-Label Study of Eculizumab in Pediatric Participants With Refractory Generalized Myasthenia Gravis (gMG) 11 Nov 2023
NCT: national clinical trial.
a Denotes industry-sponsored or cosponsored trial.

REFERENCES

  1. Punga AR, Maddison P, Heckmann JM, et al. Epidemiology, diagnostics, and biomarkers of autoimmune neuromuscular junction disorders. Lancet Neurol. Feb 2022; 21(2): 176-188. PMID 35065040
  2. Vincent A, McConville J, Farrugia ME, et al. Antibodies in myasthenia gravis and related disorders. Ann N Y Acad Sci. Sep 2003; 998: 324-35. PMID 14592891
  3. Rødgaard A, Nielsen FC, Djurup R, et al. Acetylcholine receptor antibody in myasthenia gravis: predominance of IgG subclasses 1 and 3. Clin Exp Immunol. Jan 1987; 67(1): 82-8. PMID 3621677
  4. McConville J, Farrugia ME, Beeson D, et al. Detection and characterization of MuSK antibodies in seronegative myasthenia gravis. Ann Neurol. Apr 2004; 55(4): 580-4. PMID 15048899
  5. Jaretzki A, Barohn RJ, Ernstoff RM, et al. Myasthenia gravis: recommendations for clinical research standards. Task Force of the Medical Scientific Advisory Board of the Myasthenia Gravis Foundation of America. Neurology. Jul 12 2000; 55(1): 16-23. PMID 10891897
  6. Phillips LH. The epidemiology of myasthenia gravis. Ann N Y Acad Sci. Sep 2003; 998: 407-12. PMID 14592908
  7. Somnier FE, Engel PJ. The occurrence of anti-titin antibodies and thymomas: a population survey of MG 1970-1999. Neurology. Jul 09 2002; 59(1): 92-8. PMID 12105313
  8. MacDonald BK, Cockerell OC, Sander JW, et al. The incidence and lifetime prevalence of neurological disorders in a prospective community-based study in the UK. Brain. Apr 2000; 123 ( Pt 4): 665-76. PMID 10733998
  9. McGrogan A, Sneddon S, de Vries CS. The incidence of myasthenia gravis: a systematic literature review. Neuroepidemiology. 2010; 34(3): 171-83. PMID 20130418
  10. Thomsen JLS, Andersen H. Outcome Measures in Clinical Trials of Patients With Myasthenia Gravis. Front Neurol. 2020; 11: 596382. PMID 33424747
  11. Howard JF, Utsugisawa K, Benatar M, et al. Safety and efficacy of eculizumab in anti-acetylcholine receptor antibody-positive refractory generalised myasthenia gravis (REGAIN): a phase 3, randomised, double-blind, placebo-controlled, multicentre study. Lancet Neurol. Dec 2017; 16(12): 976-986. PMID 29066163
  12. Prescribing Label: Soliris (eculizumab) injection, for intravenous use. Available at https://alexion.com/Documents/Soliris_USPI.pdf. Accessed on April 19, 2024.
  13. Muppidi S, Utsugisawa K, Benatar M, et al. Long-term safety and efficacy of eculizumab in generalized myasthenia gravis. Muscle Nerve. Jul 2019; 60(1): 14-24. PMID 30767274
  14. Center for Drug Evaluation and Research (FDA): Approval Package for Soliris. Available at https://www.accessdata.fda.gov/drugsatfda_docs/nda/2021/125166Orig1s422.pdf. Accessed on April 19, 2024.
  15. Vu T, Meisel A, Mantegazza R, et al. Terminal Complement Inhibitor Ravulizumab in Generalized Myasthenia Gravis. NEJM Evid. May 2022; 1(5): EVIDoa2100066. PMID 38319212
  16. Prescribing Label: Ultomiris (ravulizumab-cwvz) injection, for intravenous or subcutaneous use. Available at https://alexion.com/Documents/Ultomiris_USPI.pdf. Accessed on April 19, 2024.
  17. Meisel A, Annane D, Vu T, et al. Long-term efficacy and safety of ravulizumab in adults with anti-acetylcholine receptor antibody-positive generalized myasthenia gravis: results from the phase 3 CHAMPION MG open-label extension. J Neurol. Aug 2023; 270(8): 3862-3875. PMID 37103755
  18. Prescribing Label: Vyvgart (efgartigimod alfa-fcab) injection, for intravenous use. Available at https://www.argenx.com/product/vyvgart-prescribing-information.pdf. Accessed on April 19, 2024.
  19. Howard JF, Bril V, Vu T, et al. Safety, efficacy, and tolerability of efgartigimod in patients with generalised myasthenia gravis (ADAPT): a multicentre, randomised, placebo-controlled, phase 3 trial. Lancet Neurol. Jul 2021; 20(7): 526-536. PMID 34146511
  20. Statistical Analysis Plan for NCT04735432. Posted August 16, 2022. Available at https://classic.clinicaltrials.gov/ProvidedDocs/32/NCT04735432/SAP_001.pdf. Accessed on April 19, 2024.
  21. Explore Clinical data for Vygart Hytrulo. Available at https://www.vyvgarthcp.com/clinical-data/vyvgart-hytrulo. Accessed on April 19, 2024.
  22. Prescribing Label: Vyvgart Hytrulo (efgartigimod alfa and hyaluronidase-qvfc) injection, for subcutaneous use. Available at https://www.argenx.com/product/vyvgart-hytrulo-prescribing-information.pdf. Accessed on April 19, 2024.
  23. Prescribing label: Rystiggo (rozanolixizumab-noli) injection, for subcutaneous use. Available at https://www.ucb-usa.com/RYSTIGGO-prescribing-information.pdf. Accessed on April 19, 2024.
  24. Bril V, Drużdż A, Grosskreutz J, et al. Safety and efficacy of rozanolixizumab in patients with generalised myasthenia gravis (MycarinG): a randomised, double-blind, placebo-controlled, adaptive phase 3 study. Lancet Neurol. May 2023; 22(5): 383-394. PMID 37059507
  25. Narayanaswami P, Sanders DB, Wolfe G, et al. International Consensus Guidance for Management of Myasthenia Gravis: 2020 Update. Neurology. Jan 19 2021; 96(3): 114-122. PMID 33144515

Codes

Codes Number Description
CPT No CPT Code  
HCPCS J1300 Injection, eculizumab, 10 mg
  J1303 Injection, ravulizumab-cwvz, 10 mg
  J9332 Injection, efgartigimod alfa-fcab, 2mg
  J9333 Injection, rozanolixizumab-noli, 1 mg
  J9334 Injection, efgartigimod alfa, 2 mg and hyaluronidase-qvfc
ICD10 CM G70.00 Myasthenia gravis without (acute) exacerbation
  G70.01 Myasthenia gravis with (acute) exacerbation
ICD10 PCS XW033C6 Introduction of Eculizumab into Peripheral Vein, Percutaneous Approach, New Technology Group 6
  XW043C6 Introduction of Eculizumab into Central Vein, Percutaneous Approach, New Technology Group 6
Type of Service Pharmacy  
Place of Service Outpatient/Inpatient

Policy History

Date

Action

Description

07/17/24

Policy Review

Policy updated with literature review through April 9, 2024. Policy statement updated to clarify prior therapies required for Rystiggo.

06/19/24

Agents coverage Decision

Policy updated with coverage determination among class based on effectiveness.  Class review was discussed at the Drug Evaluation Committee of june 17, 2024.  Effectiveness of  rozanolixizumab-noli is lowest on class. Due availability of other agents rozanolixizumab-noli  is considered as an excluded therapy.  Coverage will be considered on an exception basis.

01/08/24

Annual Review

Policy updated with literature review through September 14, 2023; relevant information on rozanolixizumab and subcutaneous formulation for efgartigimod was added. Policy statements were edited. Subcutaneous formulation for efgartigimod and rozanolixizumab may be considered medically necessary for initial treatment or continued treatment of individuals with refractory myasthenia gravis who meet criteria. Title of the policy was changed from "Biological Treatments for Anti-Acetylcholine Receptor Antibody Positive Refractory Myasthenia Gravis" to "Biological Treatments for Refractory Myasthenia Gravis"Multiple references were added.

08/14/23

Annual Review

Policy updated with literature review through April 15, 2023; multiple references were added. Evidence review section was extensively revised including correction that the primary endpoint of the pivotal REGAIN trial for eculizumab met statistical significance. Policy statements for eculizumab and ravulizumab-cwvz for initial & continuation treatment were combined, continuation of treatment for all was simplified to “continues to meet initial treatment criteria,” MG-ADL for efgartigimod alfa-fcab was updated to the score values only, and the requirements specifica to safety for efgartigimod alfa-fcab were moved from the statement to guidelines. Policy Guidelines were updated to combine BBW and ACIP since they are directly related and remove Drug-Drug Interactions as that is not standard in BCBSA Policy Guidelines. 

08/11/22

Created

New policy.  Policy created with literature review through May 13, 2022. Eculizumab, ravulizumab, and efgartigimod may be considered medically necessary for the treatment of refractory myasthenia gravis when used according to the stated criteria.