MedPath

Amifampridine Advanced Drug Monograph

Published:Aug 20, 2025

Generic Name

Amifampridine

Brand Names

Firdapse, Ruzurgi, Firdapse (previously Zenas), Amifampridine SERB

Drug Type

Small Molecule

Chemical Formula

C5H7N3

CAS Number

54-96-6

Associated Conditions

Lambert Eaton Myasthenic Syndrome (LEMS)

A Comprehensive Monograph on Amifampridine (DB11640) for the Treatment of Lambert-Eaton Myasthenic Syndrome

1.0 Executive Summary

Amifampridine is a small molecule drug classified as a voltage-gated potassium channel blocker. It stands as the first-line, evidence-based symptomatic treatment for Lambert-Eaton Myasthenic Syndrome (LEMS), a rare and debilitating autoimmune disorder of the neuromuscular junction.[1] The core pathophysiology of LEMS involves an antibody-mediated reduction in presynaptic calcium influx, which impairs the release of the neurotransmitter acetylcholine (ACh) and leads to profound muscle weakness. Amifampridine directly counteracts this deficit by prolonging the presynaptic action potential, thereby enhancing calcium entry and restoring ACh release at the neuromuscular junction.[1]

The clinical efficacy of amifampridine has been unequivocally established in pivotal Phase III, randomized, placebo-controlled withdrawal trials. These studies demonstrated that patients continuing amifampridine maintained muscle strength and functional status, whereas those withdrawn to placebo experienced rapid and statistically significant deterioration.[5] This robust evidence base supports its approval for both adult and pediatric patients aged six years and older.

The safety profile of amifampridine is well-characterized, with the most significant risk being dose-dependent seizures, which has led to a contraindication in patients with a history of seizure disorders.[4] The drug's metabolism is critically dependent on the N-acetyltransferase 2 (NAT2) enzyme, which is subject to common genetic polymorphisms. Individuals who are "slow acetylators" experience significantly higher drug exposure, increasing their risk for adverse events and necessitating a personalized dosing approach with a lower starting dose.[4]

The regulatory and commercial history of amifampridine in the United States is particularly noteworthy. The legal conflict between the manufacturers of two different formulations, Firdapse® (amifampridine phosphate) and the subsequently withdrawn Ruzurgi® (amifampridine), has become a significant case study in the interpretation and application of the Orphan Drug Act, highlighting complex tensions between incentivizing rare disease drug development and ensuring patient access and market competition.[11] Overall, amifampridine represents a targeted and effective therapy that has fundamentally improved the management of LEMS, while its development journey offers important lessons for the broader pharmaceutical landscape.

2.0 Introduction to Amifampridine and Lambert-Eaton Myasthenic Syndrome (LEMS)

2.1 The Pathophysiology and Clinical Burden of Lambert-Eaton Myasthenic Syndrome (LEMS)

Lambert-Eaton Myasthenic Syndrome (LEMS) is a rare autoimmune disorder affecting the presynaptic terminal of the neuromuscular junction.[1] Its worldwide prevalence is estimated to be between 1 in 250,000 and 1 in 333,300, or approximately 2.8 to 3.4 cases per million people, firmly classifying it as a rare disease.[14]

The fundamental pathophysiology of LEMS is an autoantibody-mediated attack on P/Q-type voltage-gated calcium channels (VGCCs) located on the presynaptic nerve terminal.[1] These channels are essential for the influx of calcium ions that triggers the release of the neurotransmitter acetylcholine (ACh). The binding of these pathogenic autoantibodies leads to a reduction in the number of functional VGCCs, which in turn causes a decrease in presynaptic calcium influx during a nerve impulse. This results in impaired quantal release of ACh into the synaptic cleft.[1] The diminished ACh signal is insufficient to reliably activate postsynaptic muscle fibers, leading to the characteristic symptoms of the disease. This impairment is objectively measured as a low baseline compound muscle action potential (CMAP) amplitude in electrodiagnostic studies.[1]

Clinically, LEMS presents with a characteristic triad of symptoms: proximal muscle weakness (affecting the hips and shoulders), autonomic dysfunction (e.g., dry mouth, constipation, erectile dysfunction), and depressed or absent deep tendon reflexes.[1] A critical aspect of LEMS is its strong association with malignancy, making it a paraneoplastic syndrome in a majority of cases. Approximately 50-60% of individuals diagnosed with LEMS have an underlying cancer, most commonly small-cell lung cancer (SCLC).[1] In many instances, the onset of neurological symptoms precedes the diagnosis of the cancer, positioning LEMS as an important clinical indicator that should prompt a thorough oncological investigation.[17]

Diagnosis is confirmed through a combination of clinical presentation, serological testing, and electrodiagnostic studies. The detection of anti-VGCC antibodies in the serum is highly specific for LEMS and is positive in approximately 85-95% of patients.[17] Electrodiagnostic testing reveals a pathognomonic finding: a low-amplitude CMAP at rest that demonstrates a remarkable incremental response (often greater than 100%) following a brief period of high-frequency repetitive nerve stimulation or maximal voluntary muscle contraction. This post-exercise facilitation distinguishes LEMS from other neuromuscular disorders like myasthenia gravis.[18]

The paraneoplastic nature of LEMS presents a dual therapeutic challenge. Management must address not only the debilitating neuromuscular symptoms but also the urgent need to screen for and treat the underlying malignancy. In this context, therapies that restore muscle function are not merely palliative; they are crucial components of supportive care. By improving a patient's strength and overall functional status, such treatments can enhance their ability to tolerate and participate in aggressive anticancer regimens like chemotherapy and radiation, which are often necessary for SCLC.[20]

2.2 Amifampridine: A Targeted Symptomatic Therapy

Amifampridine, also known by its chemical name 3,4-diaminopyridine (3,4-DAP), is a small molecule drug that serves as a targeted symptomatic therapy for LEMS.[1] Its development was specifically aimed at counteracting the functional deficit—impaired ACh release—that defines the disease. As a symptomatic treatment, amifampridine alleviates the clinical manifestations of muscle weakness by directly addressing the physiological bottleneck at the neuromuscular junction. It does not, however, alter the underlying autoimmune disease process responsible for the destruction of calcium channels.[1] Due to its proven efficacy and targeted mechanism, amifampridine is now recognized by major regulatory bodies and neurological societies as the first-line symptomatic treatment for LEMS.[2]

3.0 Physicochemical Properties and Formulations

3.1 Chemical Identity and Structure

Amifampridine is a simple pyridine derivative with two amino groups. Its precise chemical identity is well-established and cataloged across multiple chemical and pharmacological databases.

  • Systematic (IUPAC) Name: Pyridine-3,4-diamine.[4]
  • Common Synonyms: 3,4-diaminopyridine, 3,4-DAP.[1]
  • DrugBank ID: DB11640.[1]
  • CAS Number: 54-96-6 (for the free base).[4]
  • Molecular Formula: C5​H7​N3​.[4]
  • Molecular Weight: 109.13 g/mol.[4]

3.2 Physical and Chemical Characteristics

Amifampridine exists as a solid at room temperature with the following properties:

  • Appearance: Yellow to brownish crystalline powder or solid.[4]
  • Melting Point: Decomposes in the range of 216–220 °C.[4]
  • Solubility: It is readily soluble in water, with a reported solubility of 24–25 g/L at 20 °C, and is also soluble in alcohols. Its solubility in nonpolar solvents like diethyl ether is slight.[4]

3.3 Commercial Formulations: The Importance of the Phosphate Salt

While the free base form of amifampridine (3,4-DAP) was used for decades through compassionate use programs and compounding pharmacies, the commercially approved pharmaceutical products, Firdapse® and the formerly available Ruzurgi®, utilize amifampridine phosphate.[1] This phosphate salt (CAS Number: 446254-47-3) was specifically developed to enhance the pharmaceutical properties of the drug.[4] The primary advantage of the phosphate salt is its improved chemical stability, which allows the final tablet formulation to be stored at room temperature without the need for refrigeration. This enhancement in stability was a critical step in transforming amifampridine from an investigational compound into a regulated, commercially viable medicine with a reliable shelf-life and consistent manufacturing standards.[4]

The development of the phosphate salt was not intended to alter the therapeutic action of the drug but rather to address the practical requirements of pharmaceutical production and distribution. This transition underscores a crucial aspect of drug development: pharmaceutical science innovations, such as salt form selection to improve stability, are often as vital as clinical discoveries in bringing a therapy to patients on a global scale. Each 10 mg tablet of the approved formulation contains 18.98 mg of amifampridine phosphate, which is stoichiometrically equivalent to 10 mg of the active amifampridine free base moiety.[25]

Table 1: Key Chemical and Physical Identifiers for Amifampridine

Property/IdentifierValueSource(s)
IUPAC Namepyridine-3,4-diamine4
Common NamesAmifampridine, 3,4-diaminopyridine, 3,4-DAP1
DrugBank IDDB116401
CAS Number (Base)54-96-64
CAS Number (Phosphate Salt)446254-47-34
UNII (Base)RU4S6E2G0J4
Molecular FormulaC5​H7​N3​4
Molecular Weight109.13 g/mol4
AppearanceYellow to brownish crystalline solid4
Melting Point216-220 °C (decomposes)4
Water Solubility24-25 g/L (at 20 °C)4
SMILESc1cncc(c1N)N4
InChIInChI=1S/C5H7N3/c6-4-1-2-8-3-5(4)7/h1-3H,7H2,(H2,6,8)4
InChIKeyOYTKINVCDFNREN-UHFFFAOYSA-N4

4.0 Clinical Pharmacology

4.1 Mechanism of Action

Amifampridine functions as a broad-spectrum, non-specific blocker of voltage-gated potassium channels (VGKCs), which are crucial for the repolarization phase of the neuronal action potential.[1] Its primary therapeutic target is the population of fast VGKCs located on the presynaptic nerve terminal, including the Kv1.1 subtype (Potassium voltage-gated channel subfamily A member 1).[1]

The mechanism proceeds through a clear sequence of events:

  1. Amifampridine binds to and blocks the potassium channels, inhibiting the efflux of potassium ions (K+) from the nerve terminal that would normally occur at the peak of an action potential.[1]
  2. This blockade of potassium efflux slows or prevents the repolarization of the cell membrane, thereby prolonging the duration of the depolarized state.[1]
  3. The prolonged depolarization, in turn, increases the open-state probability of the voltage-gated calcium channels (VGCCs), keeping them open for a longer period.[1]
  4. This extended "open time" for VGCCs augments the total influx of calcium ions (Ca2+) into the presynaptic nerve ending.[1]
  5. The resulting increase in intracellular calcium concentration enhances the exocytosis of synaptic vesicles containing acetylcholine (ACh), leading to a greater release of the neurotransmitter into the synaptic cleft for a given nerve impulse.[1]

This mechanism represents a direct and elegant pharmacological countermeasure to the core pathophysiology of LEMS. The autoimmune disease reduces the number of functional VGCCs, thereby lowering the probability of ACh release. Amifampridine does not repair this damage but instead increases the duration of the opportunity for calcium influx through the remaining functional channels. In doing so, it effectively bypasses the functional bottleneck created by the disease, compensating for the reduced channel density and restoring a more normal level of neurotransmitter release.

4.2 Pharmacodynamics

The primary pharmacodynamic effect of amifampridine is the potentiation of neuromuscular transmission, which translates directly into improved muscle function for patients with LEMS.[1] This effect can be objectively and quantitatively measured through electrodiagnostic studies. A key biomarker of amifampridine's activity is the improvement in the amplitude of the compound muscle action potential (CMAP), which is characteristically low in LEMS patients at rest.[1] Clinical studies have consistently demonstrated that treatment with amifampridine leads to statistically and clinically significant increases in CMAP amplitudes, reflecting the restoration of more effective communication between nerve and muscle.[31]

Beyond its primary site of action at the neuromuscular junction, amifampridine's mechanism as a potassium channel blocker can influence other neuronal pathways. It has been shown to potentiate both cholinergic and adrenergic transmission in other parts of the nervous system. For example, it can stimulate the release of dopamine and noradrenaline in the brain and may affect neurotransmission in the gastrointestinal tract, which could contribute to both therapeutic effects (e.g., improvement in autonomic symptoms like dry mouth) and some of its side effects.[1]

5.0 Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)

5.1 Absorption and Bioavailability

Following oral administration, amifampridine is absorbed rapidly and almost completely from the gastrointestinal tract, with a bioavailability reported to be between 93% and 100%.[4] Peak plasma concentrations (

Tmax​) are achieved quickly, typically within 0.6 hours (approximately 36 minutes) in the fasted state, indicating a rapid onset of action.[4]

The presence of food affects the rate, but not the overall extent, of absorption. Administration with a high-fat meal delays the time to peak concentration to approximately 1.3 hours and reduces the peak concentration (Cmax​) by about 44%. However, the total drug exposure, as measured by the area under the curve (AUC), is not significantly altered by food intake.[4]

5.2 Distribution

Amifampridine exhibits very low binding to plasma proteins, with a high unbound fraction in the plasma.[26] This characteristic allows the drug to distribute readily and extensively from the bloodstream into body tissues. Studies have shown that tissue concentrations of amifampridine are generally similar to or even greater than the concentrations found in plasma, indicating effective penetration to its sites of action.[1]

5.3 Metabolism and Pharmacogenomic Considerations

Amifampridine is primarily cleared from the body through metabolism. It is deactivated via N-acetylation to a single, pharmacologically inactive metabolite, 3-N-acetylamifampridine.[4] This metabolic transformation is catalyzed by N-acetyltransferase enzymes, with N-acetyltransferase 2 (NAT2) playing the predominant role.[4]

The gene encoding the NAT2 enzyme is highly polymorphic in the human population. These genetic variations result in distinct phenotypes with different enzyme activity levels, commonly categorized as slow, intermediate, and fast acetylators.[9] This genetic variability is the single most important factor driving the wide inter-individual differences observed in amifampridine's pharmacokinetics.

Individuals who are "slow acetylators" (also known as NAT2 poor metabolizers) metabolize the drug at a much slower rate. This leads to a significant accumulation of the parent drug, resulting in substantially higher systemic exposure—with AUC values up to nine times higher—and a prolonged elimination half-life compared to "fast acetylators".[4] This markedly increased exposure in slow acetylators directly correlates with a higher incidence and intensity of dose-dependent adverse reactions, such as paresthesias, nausea, and headache.[1]

This direct and clinically significant gene-drug interaction makes the clinical pharmacology of amifampridine a textbook example of the importance of pharmacogenomics. The clear causal pathway—from NAT2 genotype to enzyme activity, to the rate of metabolism, to systemic drug exposure, and ultimately to the risk of adverse events—necessitates a personalized medicine approach. In recognition of this, regulatory authorities recommend initiating treatment at the lowest possible starting dose for patients who are known to be NAT2 poor metabolizers, in order to mitigate the risk of toxicity.[7]

5.4 Excretion

Amifampridine and its inactive acetylated metabolite are eliminated from the body primarily through renal excretion into the urine.[26] Following a single oral dose, approximately 19% of the drug is excreted unchanged in the urine, while the vast majority (74% to 82%) is eliminated as the 3-N-acetylamifampridine metabolite.[26] The biological half-life of amifampridine is approximately 2.5 hours, but this value exhibits significant variability among individuals, a phenomenon largely attributable to the differences in metabolic rate dictated by their NAT2 acetylator status.[4]

6.0 Clinical Efficacy in Lambert-Eaton Myasthenic Syndrome

6.1 Evidence from Pivotal Phase III Clinical Trials

The clinical efficacy of amifampridine phosphate (Firdapse®) for the treatment of LEMS was definitively established in two pivotal Phase III, multicenter, randomized, double-blind, placebo-controlled withdrawal studies conducted in adult patients.[5] The choice of a randomized withdrawal design was a methodologically sound and efficient strategy for demonstrating the drug's efficacy in a rare disease population. Enrolling treatment-naïve patients in a traditional placebo-controlled trial for a debilitating condition with a known effective therapy can be ethically challenging and logistically slow due to the small patient pool. The withdrawal design leverages a population of existing users, provides all participants with active drug during an initial open-label phase, and can demonstrate a clear, causal link between the drug and clinical stability in a shorter timeframe. The rapid and significant deterioration observed upon withdrawal to placebo provides powerful evidence that the drug is responsible for maintaining the therapeutic benefit.

  • Study 1 (LMS-002, NCT01377922): This study enrolled 38 adult LEMS patients who were stable on amifampridine treatment. Following an open-label run-in phase, patients were randomized to either continue their stable dose of amifampridine or undergo a tapered withdrawal to placebo over a 14-day double-blind period. The results clearly demonstrated that patients who were withdrawn to placebo experienced a statistically significant worsening of their condition compared to those who remained on active treatment.[7]
  • Study 2 (LMS-003, NCT02970162): This trial served as a confirmatory study. It employed a similar withdrawal design but over a shorter 4-day period in 26 stable LEMS patients. The results corroborated the findings of the first study, showing a rapid and significant clinical deterioration in patients who were switched to placebo compared to those who continued on amifampridine.[6]

6.2 Analysis of Primary and Secondary Efficacy Endpoints

To provide a comprehensive assessment of the drug's benefit, both pivotal trials utilized two co-primary endpoints that captured both objective, physician-assessed changes and subjective, patient-reported outcomes.

  • Co-Primary Endpoints:
  1. Quantitative Myasthenia Gravis (QMG) Score: This is a 13-item, physician-rated scale that objectively measures muscle strength across different body regions. In both studies, patients randomized to the placebo group showed a statistically significant worsening (an increase) in their QMG scores, indicating a decline in muscle strength.[6]
  2. Subject Global Impression (SGI): This is a 7-point, patient-reported scale where individuals rate their overall sense of physical well-being. Patients in the placebo groups reported a statistically significant decline in their global impression, confirming that the objective muscle weakness was accompanied by a perceived negative impact on their daily lives.[6]
  • Key Secondary Endpoint:
  • Clinical Global Impression-Improvement (CGI-I): This is a 7-point, physician-rated scale assessing the change in a patient's overall condition. This endpoint also demonstrated a significant worsening in the placebo groups, aligning the clinicians' assessments with the objective QMG scores and the patients' self-reports.[6]

Collectively, the robust and consistent results from these two pivotal trials provided Class I evidence for the efficacy of amifampridine as a symptomatic treatment for LEMS, forming the foundation of its regulatory approvals worldwide.[6]

6.3 Efficacy in the Pediatric Population

The indication for Firdapse® was expanded by the U.S. Food and Drug Administration (FDA) to include pediatric patients aged six years and older. This approval was based on the extrapolation of efficacy demonstrated in the adult trials, supported by safety and pharmacokinetic data collected from pediatric patients in an expanded access program. This approach is common for rare diseases where conducting separate, large-scale pediatric efficacy trials is not feasible.[7]

Table 2: Summary of Pivotal Phase III Clinical Trial Results for Amifampridine in LEMS

ParameterStudy 1 (LMS-002 / NCT01377922)Study 2 (LMS-003 / NCT02970162)
Study DesignRandomized, double-blind, placebo-controlled withdrawalRandomized, double-blind, placebo-controlled withdrawal
Number of Patients (N)38 (Firdapse® N=16, Placebo N=22)26 (Firdapse® N=13, Placebo N=13)
Double-Blind Duration14 days4 days
Primary Endpoint 1: Change in QMG Score (LS Mean)
Firdapse® Arm+0.1-0.04
Placebo Arm+1.8+6.50
Treatment Difference (Firdapse - Placebo)-1.7 (95% CI: -3.4, -0.0)-6.54 (95% CI: -9.78, -3.29)
p-valuep=0.045p=0.0004
Primary Endpoint 2: Change in SGI Score (LS Mean)
Firdapse® Arm-1.1-2.15
Placebo Arm+0.7+0.80
Treatment Difference (Firdapse - Placebo)1.8 (95% CI: 0.7, 3.0)2.95 (95% CI: 1.53, 4.38)
p-valuep=0.003p=0.0003
Secondary Endpoint: Change in CGI-I Score (Mean)
Treatment Difference (Firdapse - Placebo)-1.1-2.7
p-valuep=0.02p=0.002
Source(s)55

Note: For QMG and CGI-I scores, a negative treatment difference favors Firdapse® (less worsening). For the SGI score, a positive treatment difference favors Firdapse® (less worsening).

7.0 Safety and Tolerability Profile

7.1 Adverse Event Profile

The safety profile of amifampridine is well-defined, with most adverse reactions being dose-dependent and related to its mechanism of action.

  • Most Common Adverse Reactions (>10%): The adverse events reported most frequently in clinical trials include paresthesias (sensations of tingling or numbness, particularly around the mouth and in the fingers and toes), upper respiratory tract infection, abdominal pain, nausea, diarrhea, headache, elevated liver enzymes, back pain, hypertension, and muscle spasms.[7] Paresthesias are a very common and characteristic side effect, often considered an on-target effect resulting from the drug's enhancement of sensory nerve excitability.[7]

The safety profile of amifampridine is intrinsically linked to its fundamental pharmacology. The same potassium channel blockade that produces the desired therapeutic effect at the neuromuscular junction—enhanced neuronal transmission—can also lead to generalized neuronal hyperexcitability in other parts of the nervous system. This manifests as the most common adverse event, paresthesia, and, at higher exposures, as the most serious adverse event: seizures. This illustrates a classic pharmacological principle where the on-target effect is responsible for both efficacy and dose-limiting toxicity. This understanding is key to managing the drug's risks through careful dose titration and avoiding concomitant medications that could further increase neuronal excitability.

7.2 Warnings, Precautions, and Contraindications

The use of amifampridine is associated with several significant risks that require careful patient selection and monitoring.

  • Seizures (Most Serious Risk): Amifampridine is known to lower the seizure threshold and carries a significant, dose-dependent risk of inducing seizures.[7] Seizures have been reported in patients both with and without a prior history of epilepsy. This risk forms the basis of the primary contraindication for the drug.
  • Contraindication: Amifampridine is strictly contraindicated in patients with a history of seizures.[5] Treatment should be discontinued if a seizure occurs.
  • Hypersensitivity: The drug is contraindicated in patients with a known hypersensitivity to amifampridine or any other aminopyridine compound. Although rare, serious allergic reactions, including anaphylaxis, are a potential risk.[7]
  • QT Prolongation: Due to its effects on ion channels, there is a theoretical risk that amifampridine could prolong the QT interval of the electrocardiogram (ECG), which can predispose patients to serious cardiac arrhythmias.
  • Contraindication: Concomitant use with other medications known to prolong the QT interval (e.g., sultopride, cisapride) is contraindicated, as is its use in patients with congenital long QT syndrome.[3] Annual ECG monitoring is recommended for patients on therapy.[41]
  • Hepatotoxicity: Cases of transiently elevated liver enzymes have been reported in clinical trials. While typically mild, rare instances of more significant, biopsy-confirmed drug-induced hepatotoxicity have been documented, although these have been reversible upon discontinuation of the drug.[8]

7.3 Use in Specific Populations

  • Renal and Hepatic Impairment: Since amifampridine is cleared by the kidneys, impairment of renal or hepatic function can lead to increased drug exposure and a higher risk of adverse events. Therefore, it is recommended that amifampridine be used with caution in these populations. A lower starting dose and a slower, more gradual dose titration schedule are advised.[7]
  • Pregnancy and Lactation: There is a lack of adequate and well-controlled studies in pregnant women. Animal reproduction studies have suggested a potential for fetal harm, including an increased number of stillbirths. A pregnancy exposure registry has been established to collect data on outcomes in women exposed to amifampridine during pregnancy.[26] The drug should not be used during pregnancy or breastfeeding unless the potential benefit to the mother clearly justifies the potential risk to the fetus or infant.[41]

8.0 Clinically Significant Drug Interactions

The most clinically important drug interactions associated with amifampridine are pharmacodynamic in nature, stemming from additive effects on neuronal excitability and cholinergic systems.

8.1 Pharmacodynamic Interactions

  • Drugs that Lower the Seizure Threshold: The primary safety concern with amifampridine is its potential to cause seizures. Co-administration with other medications that are also known to lower the seizure threshold can have an additive effect, significantly increasing this risk. Such drugs include bupropion, tramadol, certain antipsychotics (e.g., clozapine), and tricyclic antidepressants (e.g., amitriptyline). The decision to use amifampridine concurrently with these agents must be made with extreme caution, and in many cases, the combination should be avoided.[7]
  • Drugs with Cholinergic Effects: Amifampridine enhances cholinergic transmission by increasing ACh release. When used concomitantly with other drugs that have cholinergic effects, such as direct or indirect cholinesterase inhibitors (e.g., pyridostigmine, donepezil), there is a potential for potentiated cholinergic activity. This can increase the risk of cholinergic adverse reactions (e.g., nausea, diarrhea, abdominal cramps, increased salivation).[7]
  • Drugs that Prolong the QT Interval: As noted in the safety section, concomitant use of amifampridine with other drugs known to prolong the QT interval is contraindicated. This is due to the potential for an additive effect on cardiac repolarization, which could increase the risk of life-threatening arrhythmias like Torsades de Pointes.[3]

8.2 Pharmacokinetic Interactions

Amifampridine's metabolic pathway is relatively simple, which limits its potential for certain types of pharmacokinetic interactions. It is not a significant substrate, inhibitor, or inducer of the major Cytochrome P450 (CYP) enzyme families that are responsible for the metabolism of many other drugs. This suggests a low likelihood of CYP-mediated drug-drug interactions.[26] The clinical focus for interaction management should therefore remain on the pharmacodynamic effects.

Table 3: Clinically Significant Drug Interactions with Amifampridine

Interacting Drug Class/AgentNature of InteractionClinical Recommendation/ManagementSource(s)
Drugs that Lower Seizure Threshold (e.g., Bupropion, Tramadol, Antipsychotics, TCAs)Additive pharmacodynamic effect, increasing the risk of seizures.Carefully consider the risk-benefit profile. Avoid combination if possible or use with extreme caution and close monitoring.7
Drugs with Cholinergic Effects (e.g., Pyridostigmine, Donepezil)Potentiation of cholinergic effects, increasing the risk of cholinergic adverse reactions.Monitor for increased cholinergic side effects (e.g., nausea, diarrhea, cramping). Dose adjustment of one or both agents may be necessary.7
Drugs that Prolong the QT Interval (e.g., Sultopride, Cisapride, certain antiarrhythmics and antipsychotics)Additive effect on cardiac repolarization, increasing the risk of serious cardiac arrhythmias.Contraindicated. Concomitant use should be avoided.3
Non-depolarising Muscle Relaxants (e.g., Mivacurium)Potential for decreased effect of the muscle relaxant due to enhanced cholinergic transmission.Consider the potential for interaction when used in a surgical or procedural setting.27

9.0 Dosage, Administration, and Titration

The dosing and administration guidelines for amifampridine are meticulously designed to mitigate its primary safety risk: dose-dependent seizures. The core principles of the regimen—"start low, go slow," divided daily dosing, and strict maximum single-dose limits—are all strategies to manage and minimize peak plasma concentrations (Cmax​), which are the most likely trigger for neuronal hyperexcitability. The slow titration allows the patient's body to acclimate and helps the clinician identify the lowest effective dose, while dividing the total daily dose into three or four administrations smooths the plasma concentration curve over a 24-hour period, preventing the high peaks that would occur with less frequent dosing. The explicit cap on the maximum single dose is a direct measure to prevent acute toxicity.

9.1 Recommended Dosing Regimens (Firdapse®)

  • Adults and Pediatric Patients (weighing 45 kg or more):
  • Starting Dosage: The recommended starting dosage is 15 mg to 30 mg per day, administered orally in 3 to 4 divided doses.
  • Titration: The total daily dosage can be increased by 5 mg every 3 to 4 days, based on clinical response and tolerability.
  • Maximum Single Dose: No single dose should exceed 20 mg.
  • Maximum Total Daily Dose: The total daily dosage should not exceed 80 mg. In June 2024, the FDA approved an increase in the maximum daily dose to 100 mg for adult patients.[7]
  • Pediatric Patients (weighing less than 45 kg):
  • Starting Dosage: The recommended starting dosage is 5 mg to 15 mg per day, administered orally in divided doses.
  • Titration: The total daily dosage can be increased by 2.5 mg every 3 to 4 days.
  • Maximum Single Dose: No single dose should exceed 10 mg.
  • Maximum Total Daily Dose: The total daily dosage should not exceed 40 mg.[7]
  • Administration: Amifampridine tablets can be taken with or without food. For patients who require dose adjustments of less than 5 mg, have difficulty swallowing tablets, or require administration via a feeding tube, a 1 mg/mL oral suspension can be prepared from the tablets.[7]

9.2 Dose Adjustments for Special Populations

A more conservative dosing approach is recommended for populations at higher risk of increased drug exposure. The lowest recommended starting daily dosage (i.e., 15 mg for adults and pediatrics ≥ 45 kg; 5 mg for pediatrics < 45 kg) should be used for:

  • Patients with any degree of renal impairment.
  • Patients with any degree of hepatic impairment.
  • Patients who are known to be NAT2 poor metabolizers.

In these specific populations, dose titration should be performed more slowly and with heightened clinical monitoring for adverse reactions.[7]

Table 4: Recommended Dosage and Administration for Firdapse® (Amifampridine)

Patient PopulationRecommended Starting Daily DosageTitration ScheduleMaximum Single DoseMaximum Total Daily Dose
Adults (any weight)15 mg to 30 mg (in 3-4 divided doses)Increase by 5 mg daily every 3-4 days20 mg100 mg
Pediatrics (≥ 45 kg)15 mg to 30 mg (in 3-4 divided doses)Increase by 5 mg daily every 3-4 days20 mg80 mg
Pediatrics (< 45 kg)5 mg to 15 mg (in 3-4 divided doses)Increase by 2.5 mg daily every 3-4 days10 mg40 mg
Patients with Renal/Hepatic Impairment or Known NAT2 Poor Metabolizers (≥ 45 kg)15 mg (in divided doses)Titrate more slowly with caution20 mg80 mg (100 mg for adults)
Patients with Renal/Hepatic Impairment or Known NAT2 Poor Metabolizers (< 45 kg)5 mg (in divided doses)Titrate more slowly with caution10 mg40 mg
Source(s)7

10.0 Regulatory and Commercial Landscape

10.1 Regulatory Journey in the United States (FDA)

The path to regulatory approval for amifampridine in the U.S. was lengthy and complex, involving multiple companies and significant legal challenges.

  • Firdapse® (Catalyst Pharmaceuticals):
  • Amifampridine received Orphan Drug Designation from the FDA for the treatment of LEMS on November 12, 2009.[47]
  • Catalyst Pharmaceuticals' development program faced a setback in February 2016 when the FDA issued a "Refusal to File" letter for its initial New Drug Application (NDA).[40]
  • After addressing the FDA's concerns, the company resubmitted the NDA in March 2018. The application was granted Priority Review status.[40]
  • On November 28, 2018, the FDA approved Firdapse® for the treatment of LEMS in adults.[11]
  • The indication was expanded to include pediatric patients (aged 6 years and older) on September 29, 2022.[38]
  • In June 2024, the FDA approved a supplemental NDA to increase the maximum recommended daily dose for adults from 80 mg to 100 mg.[4]
  • Ruzurgi® (Jacobus Pharmaceutical):
  • On May 6, 2019, the FDA approved Ruzurgi®, an amifampridine formulation from Jacobus Pharmaceutical, specifically for the treatment of LEMS in pediatric patients aged 6 to less than 17 years.[12]

10.2 Approval and Marketing in Europe (EMA) and Other Regions

The regulatory process in Europe preceded that in the U.S. by nearly a decade.

  • The European Medicines Agency (EMA) granted amifampridine an Orphan Medicine designation for LEMS on December 18, 2002.[3]
  • On December 23, 2009, the European Commission granted a marketing authorization for the drug under the brand name Zenas. The name was subsequently changed to Firdapse® on January 28, 2010.[3]
  • Firdapse® became commercially available in the European Union in April 2010.[50]
  • The drug has also been approved by Health Canada (July 31, 2020) and is currently under regulatory review in Japan.[51]

10.3 The Firdapse® vs. Ruzurgi® Legal Case: A Landmark in Orphan Drug Exclusivity

The sequential approvals of Firdapse® and Ruzurgi® in the U.S. market sparked a significant legal and commercial controversy that has become a case study in the application of the Orphan Drug Act.

  • Upon its approval in 2018, Firdapse® was granted a seven-year period of orphan drug exclusivity (ODE) for the treatment of LEMS in adults.[13]
  • The FDA's subsequent approval of Ruzurgi® in 2019 for a pediatric LEMS population was immediately challenged by Catalyst Pharmaceuticals. Catalyst filed a lawsuit against the FDA, arguing that the approval of Ruzurgi® for any LEMS indication violated Firdapse's broader market exclusivity for the disease.[12]
  • The legal battle was highly contentious, particularly because Jacobus Pharmaceutical had provided its amifampridine formulation to patients for free for many years through compassionate use programs, and Ruzurgi® was expected to be a much lower-cost alternative to Firdapse®, which was launched with an annual list price of $375,000.[4]
  • Ultimately, the U.S. Court of Appeals sided with Catalyst. In compliance with the court's ruling, the FDA officially withdrew its approval for Ruzurgi® in February 2022.[12] Following this, Catalyst's Firdapse® received an expanded indication to cover the pediatric population, solidifying its position as the sole FDA-approved amifampridine product for LEMS in the United States.

This case illustrates the powerful and sometimes controversial consequences of the Orphan Drug Act. While the Act is designed to incentivize pharmaceutical companies to invest in developing treatments for rare diseases, this instance highlights how its exclusivity provisions can be leveraged to create a market monopoly, limit competition, and generate significant debate around drug pricing and patient access. It raises fundamental policy questions about the intended balance between rewarding innovation and ensuring the availability of affordable medicines for vulnerable patient populations.

11.0 Comparative Therapeutic Assessment

The treatment landscape for LEMS includes symptomatic therapies and disease-modifying immunomodulatory treatments. Amifampridine's position within this landscape is well-defined as the primary symptomatic agent.

11.1 Amifampridine versus Pyridostigmine

  • Mechanism of Action: The two primary symptomatic agents for LEMS have distinct mechanisms. Amifampridine works presynaptically to increase the release of ACh from the nerve terminal.[1] In contrast, pyridostigmine is a cholinesterase inhibitor that works postsynaptically in the synaptic cleft, prolonging the action of ACh by preventing its enzymatic breakdown.[20]
  • Comparative Efficacy: In a double-blind, randomized, controlled trial directly comparing the two agents in LEMS patients, amifampridine was found to be consistently and significantly superior. Amifampridine produced significant improvements in both muscle strength and CMAP amplitudes, whereas pyridostigmine was generally not effective as a long-term monotherapy for LEMS.[20]
  • Clinical Role: Based on this evidence, amifampridine is the undisputed first-line symptomatic therapy for LEMS. Pyridostigmine has a limited role and may occasionally be used as an adjunctive therapy to supplement the effects of amifampridine in some patients, but it is not considered an effective substitute.[8]

11.2 The Role of Amifampridine in Relation to Immunomodulatory Therapies

A crucial distinction in LEMS management is between symptomatic and disease-modifying therapies.

  • Therapeutic Classes: Amifampridine is a symptomatic therapy, designed to improve function by overcoming the immediate physiological deficit. Immunomodulatory treatments—such as intravenous immunoglobulin (IVIG), plasma exchange, corticosteroids (e.g., prednisone), and immunosuppressants (e.g., azathioprine)—are disease-modifying therapies that target the underlying autoimmune process by suppressing or modulating the immune system's attack on VGCCs.[1]
  • Place in Therapy: A clear, tiered treatment algorithm exists for LEMS. Amifampridine is the foundational, first-line therapy recommended for managing the symptoms of muscle weakness in all patients.[2] Immunomodulatory therapies like IVIG are generally reserved for the next tier of treatment. They are recommended for patients who have an insufficient or partial response to symptomatic treatment with amifampridine alone, or for managing acute, severe exacerbations ("flares") of the disease.[20]

Case reports and clinical consensus statements support this complementary relationship. IVIG has been shown to be effective in patients who remain significantly impaired despite optimal amifampridine therapy, indicating that the two approaches address different aspects of the disease.[59] The clinical workflow is logical: first, establish symptomatic control and improve daily function with amifampridine; then, if the underlying disease activity remains high or the symptomatic response is inadequate, introduce an immunomodulatory agent to reduce the autoimmune attack. These treatments are not mutually exclusive but are key components of a comprehensive, multi-faceted management strategy.

12.0 Conclusion and Future Perspectives

12.1 Synthesis of Clinical Value and Risk-Benefit Profile

Amifampridine has fundamentally transformed the management of Lambert-Eaton Myasthenic Syndrome. Supported by high-quality, Class I clinical evidence, it is the undisputed standard of care for the symptomatic treatment of this rare and debilitating neuromuscular disorder in both adult and pediatric populations. Its mechanism of action as a potassium channel blocker provides a direct and effective pharmacological solution to the core pathophysiological deficit of impaired acetylcholine release. The clinical benefits, including significant improvements in muscle strength and patient-reported well-being, are well-established.

The drug's risk-benefit profile is strongly positive when used appropriately. Its primary risks, most notably a dose-dependent potential for seizures, are well-characterized and can be effectively managed through adherence to contraindications (i.e., avoiding use in patients with a history of seizures), careful dose selection and titration, and vigilance for clinically significant drug interactions. The critical influence of NAT2 pharmacogenomics on drug exposure and safety underscores the importance of a personalized approach to dosing.

12.2 Unmet Needs and Future Research Directions

Despite its success, several areas for future investigation remain. Long-term safety data continues to be important, particularly to monitor for any potential clinical signals related to the theoretical carcinogenicity risk (Schwannomas) that was identified in preclinical animal studies.[41]

The targeted mechanism of amifampridine suggests potential therapeutic utility in other rare presynaptic neuromuscular disorders, such as specific subtypes of congenital myasthenic syndromes (CMS). Investigational programs exploring these indications are ongoing and represent a promising avenue for expanding the drug's clinical application.[4]

Finally, the global expansion of access to an approved, quality-controlled formulation of amifampridine remains a key objective, with regulatory submissions currently under review in additional markets such as Japan.[52] The legal and commercial history of amifampridine in the United States will continue to serve as an influential case study, shaping ongoing policy discussions about the implementation of the Orphan Drug Act and the balance between fostering innovation and ensuring affordable patient access to essential medicines for rare diseases.

Works cited

  1. Amifampridine: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 20, 2025, https://go.drugbank.com/drugs/DB11640
  2. Amifampridine - Catalyst Pharmaceuticals, accessed August 20, 2025, https://catalystpharma.com/amifampridine-phosphate-2/
  3. Firdapse (previously Zenas) | European Medicines Agency (EMA), accessed August 20, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/firdapse
  4. Amifampridine - Wikipedia, accessed August 20, 2025, https://en.wikipedia.org/wiki/Amifampridine
  5. firdapse is the recommended first-line treatment for lambert-eaton myasthenic syndrome (lems) 1, accessed August 20, 2025, https://firdapsehcp.com/about-firdapse/
  6. Amifampridine Phosphate (Firdapse) Is Effective in a Confirmatory ..., accessed August 20, 2025, https://pubmed.ncbi.nlm.nih.gov/30801481/
  7. FIRDAPSE® (amifampridine) tablets, for oral use - accessdata.fda.gov, accessed August 20, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/208078s008lbl.pdf
  8. Amifampridines are the Most Effective Drugs for Treating Lambert-Eaton Myasthenic Syndrome With a Focus on Pediatric Lambert-Eaton Myasthenic Syndrome - PMC, accessed August 20, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11220352/
  9. 3,4-Diaminopyridine | C5H7N3 | CID 5918 - PubChem, accessed August 20, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Amifampridine
  10. Amifampridine - LiverTox - NCBI Bookshelf, accessed August 20, 2025, https://www.ncbi.nlm.nih.gov/books/NBK547851/
  11. FDA Approves Firdapse® (amifampridine) for the Treatment of ..., accessed August 20, 2025, https://ir.catalystpharma.com/news-releases/news-release-details/fda-approves-firdapser-amifampridine-treatment-lambert-eaton/
  12. Ruzurgi (amifampridine) FDA Approval History - Drugs.com, accessed August 20, 2025, https://www.drugs.com/history/ruzurgi.html
  13. Amifampridine Products Prior Authorization Policy, accessed August 20, 2025, https://static.cigna.com/assets/chcp/pdf/coveragePolicies/cnf/cnf_301_coveragepositioncriteria_amifampridine_products_firdapse_ruzurgi_pa.pdf
  14. www.orpha.net, accessed August 20, 2025, https://www.orpha.net/en/disease/detail/43393#:~:text=The%20prevalence%20is%20estimated%20to,disturbance%2C%20and%20depressed%20tendon%20reflexes.
  15. Lambert-Eaton myasthenic syndrome - Orphanet, accessed August 20, 2025, https://www.orpha.net/en/disease/detail/43393
  16. Lambert–Eaton myasthenic syndrome - Wikipedia, accessed August 20, 2025, https://en.wikipedia.org/wiki/Lambert%E2%80%93Eaton_myasthenic_syndrome
  17. Lambert-Eaton Myasthenic Syndrome - Symptoms, Causes, Treatment | NORD, accessed August 20, 2025, https://rarediseases.org/rare-diseases/lambert-eaton-myasthenic-syndrome/
  18. Lambert-Eaton Myasthenic Syndrome - StatPearls - NCBI Bookshelf, accessed August 20, 2025, https://www.ncbi.nlm.nih.gov/books/NBK507891/
  19. Study Finds Similar Rates of Lambert-Eaton Myasthenic Syndrome (LEMS) in Europe and the US - Patient Worthy, accessed August 20, 2025, https://patientworthy.com/2019/05/06/study-similar-rates-lambert-eaton-myasthenic-syndrome/
  20. Treating LEMS - FIRDAPSE® (amifampridine), accessed August 20, 2025, https://firdapsehcp.com/about-lems/treating-lems/
  21. 3,4-Diaminopyridine | 54-96-6 - ChemicalBook, accessed August 20, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB6676074.htm
  22. Amifampridine | CAS#54-96-6 | 446254-47-3 MedKoo, accessed August 20, 2025, https://www.medkoo.com/products/8370
  23. Amifampridine | 54-96-6 - SynZeal, accessed August 20, 2025, https://www.synzeal.com/en/amifampridine-
  24. 3,4-Diaminopyridine =98 54-96-6 - Sigma-Aldrich, accessed August 20, 2025, https://www.sigmaaldrich.com/US/en/product/aldrich/d7148
  25. FAQs | FIRDAPSE® (amifampridine), accessed August 20, 2025, https://firdapse.com/resources/faqs/
  26. 208078Orig1s000 - accessdata.fda.gov, accessed August 20, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/208078Orig1s000OtherR.pdf
  27. Amifampridine SERB, INN-amifampridine, accessed August 20, 2025, https://ec.europa.eu/health/documents/community-register/2022/20220519155561/anx_155561_en.pdf
  28. Table 3, Key Characteristics of Amifampridine Phosphate - NCBI, accessed August 20, 2025, https://www.ncbi.nlm.nih.gov/books/NBK603392/table/tr82698683820664_ch01_t03/?report=objectonly
  29. en.wikipedia.org, accessed August 20, 2025, https://en.wikipedia.org/wiki/Amifampridine#:~:text=Mechanism%20of%20action,-In%20Lambert%E2%80%93Eaton&text=Amifampridine%20works%20by%20blocking%20potassium,muscle%20at%20the%20end%20plate.
  30. Amifampridine Phosphate Monograph for Professionals - Drugs.com, accessed August 20, 2025, https://www.drugs.com/monograph/amifampridine-phosphate.html
  31. Amifampridine for Lambert-Eaton myasthenic syndrome - PMC, accessed August 20, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9584795/
  32. In Confidence - Scottish Medicines Consortium, accessed August 20, 2025, https://scottishmedicines.org.uk/media/1239/amifampridine_phosphate_firdapse_final_july_2012_for_website.pdf
  33. This label may not be the latest approved by FDA. For current ..., accessed August 20, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/209321s000lbl.pdf
  34. Annotation of FDA Label for amifampridine and NAT2 - PharmGKB, accessed August 20, 2025, https://www.clinpgx.org/labelAnnotation/PA166185151
  35. Physiologically-Based Pharmacokinetic Modeling for Drug-Drug, accessed August 20, 2025, https://ouci.dntb.gov.ua/en/works/4N5xWaj7/
  36. Study Details | A Phase 3 Study of Amifampridine Phosphate in ..., accessed August 20, 2025, https://www.clinicaltrials.gov/study/NCT01377922
  37. A Phase 3 Study of Amifampridine Phosphate in Patients With Lambert Eaton Myasthenic Syndrome (LEMS) - Clinical Trials | Stanford Medicine, accessed August 20, 2025, https://clinicaltrials.stanford.edu/trials/a/NCT01377922.html
  38. News & Events | FIRDAPSE® (amifampridine), accessed August 20, 2025, https://firdapsehcp.com/access-and-support/news-events/
  39. Firdapse®, Ruzurgi® (amifampridine) - Alaska Department of Health, accessed August 20, 2025, https://health.alaska.gov/media/zcclcbld/202211-firdapse_ruzurgi_criteria_2022.pdf
  40. Firdapse (amifampridine phosphate) FDA Approval History - Drugs ..., accessed August 20, 2025, https://www.drugs.com/history/firdapse.html
  41. Firdapse, INN-amifampridine - European Medicines Agency, accessed August 20, 2025, https://www.ema.europa.eu/en/documents/product-information/firdapse-epar-product-information_en.pdf
  42. FIRDAPSE® (amifampridine): Official HCP Site, accessed August 20, 2025, https://firdapsehcp.com/
  43. Official Patient Site | FIRDAPSE® (amifampridine), accessed August 20, 2025, https://firdapse.com/
  44. Firdapse, amifampridine, accessed August 20, 2025, https://ec.europa.eu/health/documents/community-register/2014/20140116127639/anx_127639_en.pdf
  45. Firdapse (amifampridine) dosing, indications, interactions, adverse ..., accessed August 20, 2025, https://reference.medscape.com/drug/firdapse-amifampridine-1000094
  46. FIRDAPSE Welcome Kit Brochure, accessed August 20, 2025, https://firdapse.com/pdfs/firdapse-educational-brochure-updated.pdf
  47. Search Orphan Drug Designations and Approvals - FDA, accessed August 20, 2025, https://www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=295309
  48. Catalyst Pharmaceuticals Announces FDA Acceptance of NDA and Priority Review Status for Firdapse® (Amifampridine Phosphate) for Lambert-Eaton Myasthenic Syndrome, accessed August 20, 2025, https://ir.catalystpharma.com/news-releases/news-release-details/catalyst-pharmaceuticals-announces-fda-acceptance-nda-and/
  49. Ruzurgi (amifampridine) - accessdata.fda.gov, accessed August 20, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2019/209321Orig1s000ltr.pdf
  50. BioMarin Launches Firdapse in the European Union, accessed August 20, 2025, https://www.biomarin.com/news/press-releases/biomarin-launches-firdapse-in-the-european-union/
  51. Summary Basis of Decision for Firdapse - Drug and Health Products Portal, accessed August 20, 2025, https://dhpp.hpfb-dgpsa.ca/review-documents/resource/SBD00503
  52. Catalyst Pharmaceuticals Announces FIRDAPSE® New Drug Application Submitted in Japan by Partner DyDo Pharma, accessed August 20, 2025, https://ir.catalystpharma.com/news-releases/news-release-details/catalyst-pharmaceuticals-announces-firdapser-new-drug/
  53. Drugs@FDA: Drug Product RUZURGI (amifampridine) NDA 209321 - PharmGKB, accessed August 20, 2025, https://www.clinpgx.org/literature/15103442
  54. Medical Management - Lambert-Eaton Myasthenic Syndrome (LEMS) - Diseases, accessed August 20, 2025, https://www.mda.org/disease/lambert-eaton-myasthenic-syndrome/medical-management
  55. Update on Amifampridine as a Drug of Choice in Lambert-Eaton Myasthenic Syndrome, accessed August 20, 2025, https://touchneurology.com/neuromuscular-diseases/journal-articles/update-on-amifampridine-as-a-drug-of-choice-in-lambert-eaton-myasthenic-syndrome-2/
  56. www.mda.org, accessed August 20, 2025, https://www.mda.org/disease/lambert-eaton-myasthenic-syndrome/medical-management#:~:text=The%20treatment%20options%20are%20similar,(IVIG)%20and%20plasma%20exchange.
  57. Management/Treatment of Lambert-Eaton Myasthenic Syndrome - Scholars@Duke, accessed August 20, 2025, https://scholars.duke.edu/publication/1498400
  58. View of Amifampridine phosphate (Firdapse) - Canadian Journal of Health Technologies, accessed August 20, 2025, https://canjhealthtechnol.ca/index.php/cjht/article/view/SR0664r/873
  59. A Unique Case of Non-paraneoplastic Lambert-Eaton Myasthenic ..., accessed August 20, 2025, https://www.neurology.org/doi/10.1212/WNL.0000000000205521
  60. A Unique Case of Non-paraneoplastic Lambert-Eaton Myasthenic Syndrome Treated With Subcutaneous Immunoglobulin: A Case Report and Review of Literature - PubMed, accessed August 20, 2025, https://pubmed.ncbi.nlm.nih.gov/38903354/
  61. Is IVIG (Intravenous Immunoglobulin) therapy medically indicated for this patient's condition?, accessed August 20, 2025, https://www.droracle.ai/articles/212913/is-the-medication
  62. Amifampridine Approved for Marketing Phase N/A Trials for Congenital Myasthenia (CM) / Lambert Eaton Myasthenic Syndrome (LEMS) - DrugBank, accessed August 20, 2025, https://go.drugbank.com/drugs/DB11640/clinical_trials?conditions=DBCOND0053084%2CDBCOND0002587&status=approved_for_marketing

Published at: August 20, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.