A Comprehensive Pharmacological and Clinical Monograph on Rufinamide (DB06201)
1.0 Executive Summary
Rufinamide is a third-generation antiepileptic drug (AED) distinguished by a triazole derivative chemical structure that is unique among currently marketed anticonvulsants.[1] Its primary and globally approved indication is as an adjunctive therapy for the management of seizures associated with Lennox-Gastaut Syndrome (LGS), a severe and often treatment-refractory form of childhood-onset epileptic encephalopathy.[1] The principal mechanism of action for Rufinamide involves the modulation of voltage-gated sodium channels. Specifically, it prolongs the inactive state of these channels, thereby limiting the sustained, high-frequency neuronal firing that underpins seizure activity.[1] Emerging evidence also suggests potential secondary mechanisms, including inhibition of metabotropic glutamate receptor 5 (mGluR5) and stimulation of calcium-activated potassium channels, which may contribute to its distinct efficacy profile.
A defining characteristic of Rufinamide is its pharmacokinetic profile. It undergoes extensive metabolism to inactive metabolites, primarily through a non-cytochrome P450 pathway involving enzymatic hydrolysis by carboxylesterases.[1] This feature minimizes its susceptibility to interactions with many CYP450-modulating drugs, a significant advantage in the polypharmacy setting common to LGS. However, its clearance is notably inhibited by valproic acid, and it acts as a weak inducer of the CYP3A4 enzyme, which has important clinical implications, particularly for co-administered hormonal contraceptives. The drug exhibits slow oral absorption and has a moderate elimination half-life of approximately 6 to 10 hours.[1]
Clinically, Rufinamide has demonstrated robust efficacy in reducing the frequency of multiple seizure types in LGS, with a particularly pronounced effect on tonic-atonic seizures, or "drop attacks," which are a major cause of morbidity in this patient population.[8] The safety and tolerability profile is generally manageable, with the most common adverse effects being central nervous system-related, such as somnolence, dizziness, fatigue, and headache.[1] A critical safety consideration is its dose-dependent shortening of the cardiac QT interval, which leads to a strict contraindication in patients with the rare genetic condition known as Familial Short QT syndrome.[6] Rufinamide represents a valuable therapeutic option, offering a unique combination of targeted efficacy, a distinct metabolic profile, and patient-friendly formulations for the challenging management of Lennox-Gastaut Syndrome.
2.0 Chemical Identity and Physicochemical Properties
This section provides a detailed characterization of the chemical and physical properties of Rufinamide, which form the basis for its pharmacological activity, formulation development, and analytical identification.
2.1 Nomenclature and Identifiers
Rufinamide is identified by a standardized set of names and codes across chemical, pharmaceutical, and regulatory databases, ensuring unambiguous reference in clinical and research settings.
- Generic Name: The internationally recognized non-proprietary name for the active substance is Rufinamide.[1]
- Systematic (IUPAC) Name: The formal chemical name according to the International Union of Pure and Applied Chemistry (IUPAC) nomenclature is 1-[(2,6-difluorophenyl)methyl]-1H-1,2,3-triazole-4-carboxamide.[6]
- CAS Number: The unique registry number assigned by the Chemical Abstracts Service is 106308-44-5.[12]
- DrugBank ID: The accession number in the DrugBank database is DB06201.[1]
- Brand Names: The drug is marketed under the trade name Banzel® in the United States and other regions, and as Inovelon® in the European Union.[1]
- Synonyms and Developmental Codes: Throughout its development and in various international markets, it has been known by several synonyms and codes, including Rufinamida, CGP 33101, RUF 331, E 2080, and Xilep.[1]
2.2 Molecular and Structural Formulae
Rufinamide's molecular structure is central to its unique pharmacological profile, distinguishing it from all other major classes of antiepileptic drugs.
- Chemical Formula: The empirical formula for Rufinamide is C10H8F2N4O.[1]
- Molecular Weight: The average molecular mass is 238.19 g/mol (often rounded to 238.2 g/mol), and the precise monoisotopic mass is 238.066617 Da.[1]
- Chemical Structure: Rufinamide is a triazole derivative. Its structure is composed of a 1,2,3-triazole heterocyclic ring, which is substituted at the N1 position with a (2,6-difluorophenyl)methyl (i.e., a 2,6-difluorobenzyl) group and at the C4 position with a carboxamide group (-CONH2). This structure is not related to other AED classes such as hydantoins, benzodiazepines, or dibenzazepines.[1]
- Computational Identifiers: For database searching and computational chemistry applications, the structure is represented by the following identifiers:
- SMILES (Simplified Molecular-Input Line-Entry System): C1=CC(=C(C(=C1)F)CN2C=C(N=N2)C(=O)N)F.[19]
- InChIKey (International Chemical Identifier Key): POGQSBRIGCQNEG-UHFFFAOYSA-N.[12]
2.3 Physicochemical Properties
The physical characteristics of the Rufinamide drug substance influence its stability, formulation, and pharmacokinetic behavior.
- Appearance: In its pure form, Rufinamide is a white, crystalline, odorless, and neutral powder with a slightly bitter taste.[6]
- Solubility: The compound exhibits poor aqueous solubility, being classified as practically insoluble in water. It is slightly soluble in organic solvents such as tetrahydrofuran and methanol, and very slightly soluble in ethanol and acetonitrile.[6] Specific solubility data indicates it dissolves in DMSO at 10 mg/mL and in DMF at 2 mg/mL.[12]
- Stability and Storage: Rufinamide is a stable compound, with data suggesting stability for at least four years under proper storage conditions.[12] It is recommended to be stored in a refrigerator at 2-8°C under an inert atmosphere to ensure long-term integrity.[21]
The chemical and physical properties of Rufinamide are intrinsically linked to its clinical profile. Its structural novelty as a triazole derivative, distinct from other AEDs, is a foundational element that likely underlies its unique combination of efficacy, metabolism, and safety characteristics.[5] For instance, this unique scaffold may confer a specific binding affinity and orientation at its molecular targets, contributing to its particular effectiveness in the complex seizure milieu of LGS.
Furthermore, the compound's poor water solubility is a critical determinant of its pharmacokinetic behavior. A substance that is practically insoluble in water, like Rufinamide, often presents challenges for gastrointestinal absorption.[6] This property directly explains several key clinical observations: the relatively slow rate of absorption, with peak plasma concentrations not reached until 4-6 hours after administration; the non-linear absorption kinetics, where the fraction of the dose absorbed decreases as the dose increases; and the pronounced positive food effect.[1] The presence of food, particularly lipids, can enhance the dissolution and absorption of poorly soluble drugs, which is consistent with the finding that food increases Rufinamide's bioavailability by 34-56%.[6] This direct link between a fundamental physicochemical property and clinical pharmacokinetics necessitates the strict clinical recommendation to always administer Rufinamide with food to optimize absorption and ensure therapeutic consistency.[2]
Table 1: Chemical and Pharmacological Identifiers for Rufinamide
| Identifier Type | Value | Source(s) |
|---|
| Generic Name | Rufinamide | 1 |
| IUPAC Name | 1-[(2,6-difluorophenyl)methyl]-1H-1,2,3-triazole-4-carboxamide | 6 |
| CAS Number | 106308-44-5 | 12 |
| DrugBank ID | DB06201 | 1 |
| Molecular Formula | C10H8F2N4O | 1 |
| Average Molecular Weight | 238.19 g/mol | 1 |
| Key Brand Names | Banzel® (US), Inovelon® (EU) | 1 |
| Drug Class | Anticonvulsants; Miscellaneous Anticonvulsants | 1 |
3.0 Pharmacology and Mechanism of Action
The anticonvulsant effects of Rufinamide are attributed to its ability to modulate neuronal excitability through multiple molecular targets. While its primary mechanism is well-established, emerging evidence points to a more complex, multi-modal pharmacological profile that may be responsible for its specific efficacy in Lennox-Gastaut Syndrome.
3.1 Primary Mechanism: Modulation of Voltage-Gated Sodium Channels (VGSCs)
The principal and most well-characterized mechanism of action of Rufinamide is the modulation of voltage-gated sodium channel activity.[1] VGSCs are critical for the initiation and propagation of action potentials in neurons. During a seizure, neurons can fire in sustained, high-frequency bursts, a process dependent on the rapid cycling of VGSCs through their resting, open, and inactivated states.
Rufinamide exerts its effect by binding to the VGSCs and stabilizing them in the inactive state.[17] This action prolongs the time it takes for the channel to recover from inactivation and become available to open again. By slowing this recovery, Rufinamide effectively limits the ability of a neuron to sustain the high-frequency repetitive firing of action potentials that characterizes epileptiform activity.[6]
In vitro studies in cultured cortical neurons have quantified this effect, showing that Rufinamide significantly limits sustained repetitive firing with a half-maximal effective concentration (EC50) of 3.8 μM.[6] This membrane-stabilizing effect reduces overall neuronal hyperexcitability and is believed to be the primary mechanism by which Rufinamide prevents the propagation and spread of partial seizure activity.[1]
Further research has suggested some selectivity in Rufinamide's action on VGSC isoforms. It has been shown to inhibit the Nav1.1 isoform but not Nav1.2, Nav1.3, or Nav1.6 at a concentration of 100 µM.[12] Additionally, studies have demonstrated its ability to block tetrodotoxin-resistant (TTX-R) sodium channels, a subtype of VGSCs that are highly expressed in sensory neurons and implicated in pathological pain states.[13]
3.2 Secondary and Investigational Mechanisms
Beyond its primary action on VGSCs, Rufinamide appears to influence other key components of neuronal signaling, which may contribute to its broad anticonvulsant activity.
- Metabotropic Glutamate Receptor 5 (mGluR5) Inhibition: At higher concentrations, Rufinamide has been shown to act as an inhibitor of the metabotropic glutamate receptor 5 (mGluR5).[1] Glutamate is the primary excitatory neurotransmitter in the central nervous system, and its overactivity is a key factor in seizure generation. By inhibiting mGluR5, Rufinamide can dampen excitatory glutamatergic neurotransmission, providing a complementary mechanism to reduce neuronal hyperexcitability.[25]
- Stimulation of Calcium-Activated Potassium Channels (BKCa): Recent preclinical evidence has revealed that Rufinamide can directly stimulate the activity of large-conductance Ca2+-activated K+ channels, also known as BKCa or Maxi-K channels.[26] These channels are activated by membrane depolarization and increases in intracellular calcium, leading to an efflux of potassium ions (
K+). This outward flow of positive charge hyperpolarizes the neuronal membrane, making it more difficult for the neuron to reach the threshold for firing an action potential. By enhancing the activity of BKCa channels, Rufinamide may actively increase an inhibitory influence on neurons, further contributing to its overall anticonvulsant effect.[26]
- Carbonic Anhydrase Inhibition: Rufinamide has been identified as an inhibitor of the mitochondrial enzyme carbonic anhydrase VA (CAVA), with a high affinity (inhibitor constant, Ki = 343.8 nM). It demonstrates selectivity for this isoform over the more common CAI and CAII isoforms (Kis >10,000 nM).[12] This mechanism is shared with other AEDs, such as topiramate and zonisamide, and is thought to contribute to anticonvulsant effects through alterations in pH and ion gradients, although its clinical relevance for Rufinamide is not fully established.
The combination of these mechanisms suggests that Rufinamide's clinical efficacy, particularly in the pharmacoresistant environment of LGS, may stem from a synergistic, multi-target approach. While many AEDs modulate VGSCs, LGS is a complex epileptic encephalopathy involving widespread network dysfunction and multiple seizure types, often rendering single-mechanism agents insufficient. Rufinamide appears to counter neuronal hyperexcitability from several angles simultaneously: it directly limits the rate of neuronal firing (via VGSC inactivation), reduces excitatory synaptic input (via potential mGluR5 inhibition), and enhances an intrinsic inhibitory current (via BKCa channel stimulation). This multi-pronged attack on the cellular basis of seizures could explain its robust efficacy in a syndrome where other drugs fail.
Moreover, the drug's documented activity on TTX-R sodium channels provides a strong mechanistic foundation for its investigation in non-epileptic conditions. These specific channels are known to be upregulated in dorsal root ganglion neurons following nerve injury and are considered key players in the pathogenesis of neuropathic pain.[24] This molecular action provides a direct rationale for the initiation of clinical trials to explore Rufinamide's potential as an analgesic for chronic pain states, such as post-thoracotomy pain syndrome.[27] Although that particular trial was withdrawn, the underlying pharmacological principle remains valid and points to a broader potential for Rufinamide as a modulator of neuronal excitability beyond the context of epilepsy.[13]
4.0 Clinical Pharmacokinetics (ADME Profile)
The clinical utility and dosing of Rufinamide are governed by its pharmacokinetic profile, which describes its absorption, distribution, metabolism, and excretion (ADME). Rufinamide is characterized by slow absorption influenced by food, low protein binding, and a unique metabolic pathway that is independent of the cytochrome P450 system.
4.1 Absorption
- Bioavailability and Rate: Following oral administration, Rufinamide is well absorbed, with studies based on urinary excretion of the drug and its metabolites indicating that at least 85% of the dose is absorbed.[1] However, the rate of absorption is relatively slow, with the time to reach maximum plasma concentration (
Tmax) occurring between 4 and 6 hours after a dose.[3]
- Food Effect: The absorption of Rufinamide is significantly influenced by the presence of food. Administration with food has been shown to increase the extent of absorption by 34% and the peak plasma exposure (Cmax) by 56% compared to administration in a fasted state.[6] Consequently, to ensure maximal and consistent absorption, it is clinically recommended that Rufinamide always be administered with food.[2]
- Dose Proportionality: The pharmacokinetics of Rufinamide are non-linear with respect to dose. While plasma concentrations are approximately dose-proportional at lower doses (up to 1,600 mg/day), the extent of absorption decreases as the dose increases further.[1] This is likely due to saturation of absorption or limited dissolution of the poorly soluble drug at higher doses. Despite this, steady-state plasma concentrations are achieved relatively quickly, typically within two days of initiating or adjusting a dose, which is consistent with its elimination half-life.[28]
4.2 Distribution
- Protein Binding: Rufinamide exhibits low binding to plasma proteins, with only about 34% of the drug bound, predominantly to albumin (27%).[3] This low level of protein binding minimizes the risk of displacement interactions, where other highly protein-bound drugs could displace Rufinamide and increase its free, pharmacologically active concentration.
- Volume of Distribution: The drug distributes evenly between red blood cells and plasma.[6] The apparent volume of distribution (
Vd) is dependent on the dose and patient's body surface area. At a high daily dose of 3200 mg, the apparent Vd is approximately 50 L, indicating distribution into tissues beyond the plasma volume.[1]
4.3 Metabolism
- Primary Pathway and Metabolites: Rufinamide is extensively metabolized in the body, but crucially, it has no known active metabolites.[1] The primary and predominant metabolic pathway is the enzymatic hydrolysis of the carboxamide functional group to form a pharmacologically inactive carboxylic acid derivative, identified as CGP 47292.[1] This reaction is mediated by carboxylesterase enzymes and not by the cytochrome P450 system.[6]
- Cytochrome P450 (CYP) System Involvement: The metabolism of Rufinamide is not dependent on the CYP450 enzyme system, a characteristic that distinguishes it from many other AEDs.[2] This independence means that Rufinamide's clearance is not significantly affected by potent inducers or inhibitors of CYP enzymes. However, Rufinamide itself can have a mild influence on certain CYP isoforms. It is a weak inhibitor of CYP2E1 and, more importantly, a weak inducer of CYP3A4.[1]
4.4 Excretion
- Route of Elimination: The primary route of elimination for Rufinamide and its metabolites is via the kidneys. Renal excretion accounts for 85% to 91% of the total administered dose.[1]
- Excreted Forms: The vast majority of the drug is eliminated in the form of its metabolites. At least 66% of the dose is excreted in the urine as the inactive acid metabolite CGP 47292. A very small fraction, less than 2% of the dose, is excreted as unchanged Rufinamide.[1]
- Elimination Half-Life: The plasma elimination half-life (t1/2) of Rufinamide is consistent in both healthy volunteers and patients with epilepsy, ranging from approximately 6 to 10 hours.[1]
The pharmacokinetic profile of Rufinamide carries significant clinical implications. Its metabolism via non-CYP450 carboxylesterases is a key advantage, theoretically conferring a lower potential for drug-drug interactions (DDIs) compared to AEDs that are heavily reliant on the CYP system. This is particularly beneficial for patients with LGS, who are almost always on multiple medications. However, this profile is not without its own complexities. While Rufinamide is protected from being a "victim" of most CYP-based interactions, it is vulnerable to drugs that affect its specific metabolic pathway. The most notable example is valproic acid, a commonly co-prescribed AED that is known to inhibit carboxylesterase activity.[23] This inhibition significantly reduces Rufinamide clearance, leading to substantially higher plasma concentrations and necessitating a more cautious dosing strategy when the two drugs are used concurrently.[2]
Conversely, Rufinamide can act as a "perpetrator" in DDIs. Its weak induction of the CYP3A4 enzyme, while modest, is sufficient to accelerate the metabolism of certain CYP3A4 substrates.[2] This is the mechanistic basis for the clinically critical interaction with hormonal contraceptives, which are often metabolized by CYP3A4. Rufinamide can reduce the plasma concentrations of these contraceptives, potentially leading to contraceptive failure. This direct link between a subtle metabolic property and a major clinical risk mandates the strong recommendation for patients to use an additional, effective non-hormonal method of birth control while on Rufinamide therapy.[2] Therefore, a nuanced understanding is required: Rufinamide has a low potential to be affected by CYP modulators but a clinically relevant potential to affect CYP3A4 substrates.
Table 2: Summary of Pharmacokinetic Parameters of Rufinamide
| Parameter | Value | Clinical Implication/Comment |
|---|
| Bioavailability | ≥85% (with food) | Well absorbed, but absorption is non-linear and decreases at higher doses. |
| Tmax (Time to Peak) | 4–6 hours | Slow onset of peak plasma concentration and effect. |
| Food Effect | Absorption increased by 34%; Peak exposure increased by 56% | Must be administered with food to ensure consistent and maximal absorption. |
| Protein Binding | ~34% (primarily to albumin) | Low potential for displacement-based drug interactions. |
| Volume of Distribution (Vd) | ~50 L at 3200 mg/day | Distributes into tissues beyond the bloodstream. |
| Primary Metabolic Pathway | Hydrolysis by carboxylesterases to inactive metabolite (CGP 47292) | Metabolism is independent of the cytochrome P450 system. |
| CYP450 Involvement | Not a substrate; Weak inhibitor of CYP2E1; Weak inducer of CYP3A4 | Low risk of being a "victim" of CYP interactions, but can be a "perpetrator" by inducing CYP3A4. |
| Elimination Half-life (t1/2) | 6–10 hours | Allows for twice-daily dosing; steady state is reached in ~2 days. |
| Primary Route of Excretion | Renal (85-91%), primarily as metabolites | Less than 2% is excreted as unchanged drug. Pharmacokinetics are unaltered in renal impairment. |
5.0 Clinical Efficacy and Therapeutic Applications
The clinical utility of Rufinamide is primarily defined by its proven efficacy in its approved indication, with ongoing research exploring its potential in other neurological and psychiatric disorders.
5.1 Approved Indication: Lennox-Gastaut Syndrome (LGS)
Rufinamide is approved by major regulatory agencies, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), as an adjunctive (add-on) therapy for the treatment of seizures associated with Lennox-Gastaut Syndrome.[1] LGS is a severe form of childhood-onset epilepsy characterized by multiple seizure types (most notably tonic and atonic seizures), cognitive dysfunction, and a distinctive slow spike-and-wave pattern on electroencephalogram (EEG).
The foundation for Rufinamide's approval was a pivotal multinational, randomized, double-blind, placebo-controlled trial involving 138 patients, ranging in age from 4 to 37 years, with inadequately controlled LGS.[9] Participants were already being treated with one to three other AEDs. The study evaluated the efficacy of Rufinamide at a target dose of approximately 45 mg/kg/day against placebo over a 12-week treatment period.
The trial successfully met its three primary efficacy endpoints, demonstrating the statistically significant superiority of Rufinamide over placebo:
- Reduction in Total Seizure Frequency: Patients in the Rufinamide group experienced a median reduction of 32.7% in their total seizure frequency per 28 days, compared to a median reduction of only 11.7% in the placebo group.[8]
- Reduction in Tonic-Atonic Seizure Frequency: The effect was most pronounced on tonic-atonic seizures, also known as "drop attacks," which are a hallmark of LGS and a major cause of injury and morbidity. The Rufinamide group showed a median reduction of 42.5% in these seizures. In stark contrast, the placebo group experienced a median increase of 1.4%.[9]
- Improved Seizure Severity: Parents and guardians completed a global evaluation of the patient's condition, which showed a significantly greater improvement in overall seizure severity in the Rufinamide-treated group compared to the placebo group.[30]
The robust and highly significant effect on tonic-atonic seizures, in particular, established Rufinamide as a key therapeutic agent for LGS. This efficacy has been shown to be durable, with open-label extension studies confirming that the reduction in seizure frequency and overall tolerability are maintained with long-term treatment.[31] This clinical profile is defined by its pronounced efficacy against the most debilitating seizure type in LGS, positioning it as a cornerstone therapy for this specific indication rather than as a general broad-spectrum anticonvulsant. Its value lies in its ability to target the most dangerous and characteristic feature of the syndrome.
5.2 Other Seizure Types and Investigational Uses
While its primary indication is LGS, the pharmacological profile of Rufinamide has prompted investigation into its utility for other conditions.
- Partial Seizures: Although not part of its official FDA-approved labeling for LGS, several clinical trials have suggested that Rufinamide has efficacy as an adjunctive treatment for patients with drug-resistant partial-onset seizures.[1] This suggests a broader anticonvulsant spectrum than its narrow indication might imply.
- Generalized Anxiety Disorder (GAD): The potential of Rufinamide to modulate generalized neuronal hyperexcitability led to its investigation beyond epilepsy. A completed Phase 2 exploratory clinical trial (NCT00595231) was conducted to evaluate the effect of Rufinamide, under the developmental code SYN111, in patients with GAD.[32] This indicates an interest in its potential anxiolytic properties, though results have not led to further development for this indication to date.
- Neuropathic Pain: Based on its mechanism of action on TTX-R sodium channels, which are implicated in pain signaling, a Phase 2 clinical trial (NCT02095899) was initiated to assess Rufinamide's effect on chronic post-thoracotomy pain syndrome, a form of neuropathic pain.[27] This trial was ultimately withdrawn before completion, so its efficacy in this area remains unconfirmed.[27]
The exploration of Rufinamide for GAD and neuropathic pain, despite the lack of definitive positive outcomes leading to new indications, is significant. It reflects a deeper understanding of the drug's fundamental pharmacology. Pathological neuronal hyperexcitability is a common pathophysiological thread linking not only epilepsy but also anxiety disorders (within limbic circuits) and neuropathic pain (within peripheral sensory pathways). The investigation into these areas suggests that Rufinamide can be more accurately conceptualized not just as an "anticonvulsant," but as a broader "neuronal excitability modulator." This perspective highlights the trans-diagnostic potential of its mechanism of action, even if clinical or commercial factors have thus far limited its application to epilepsy.
6.0 Dosing, Administration, and Formulations
The effective and safe use of Rufinamide requires adherence to specific guidelines regarding its formulation, administration, and dose titration, which are tailored to different patient populations and concomitant medications.
6.1 Available Formulations
Rufinamide is available in formulations designed to accommodate the needs of the LGS patient population, which includes a wide range of ages and abilities.
- Oral Tablets: Film-coated tablets are available in 200 mg and 400 mg strengths.[4] A key practical feature is that these tablets can be administered whole, split in half, or crushed without compromising their integrity, which facilitates administration to patients with difficulty swallowing.[5]
- Oral Suspension: An oral suspension is available at a concentration of 40 mg/mL.[4] This liquid formulation is particularly useful for young pediatric patients and individuals who cannot take solid dosage forms. The oral suspension and the tablets have been shown to be bioequivalent on a milligram-for-milligram basis, allowing for a seamless transition between the two formulations as needed.[1]
The availability of these versatile formulations is a significant clinical advantage. LGS is a severe pediatric epilepsy, and the patient population often includes very young children (approved for use down to one year of age) and individuals with intellectual disabilities or dysphagia.[33] For these patients, swallowing whole tablets can be impossible. The provision of a bioequivalent oral suspension and the confirmation that tablets can be crushed directly addresses this common clinical challenge, simplifying medication administration for caregivers and reducing the risk of dosing errors or non-compliance.
6.2 Administration Guidelines
To ensure optimal pharmacokinetic performance, the following administration guidelines should be followed:
- Administration with Food: Rufinamide must be administered with food. As established in pharmacokinetic studies, food significantly enhances its absorption and bioavailability.[2]
- Dosing Frequency: The total calculated daily dose should be divided into two equal doses, administered approximately 12 hours apart (e.g., morning and evening).[17]
6.3 Dosing and Titration in Lennox-Gastaut Syndrome
The dosing for Rufinamide is weight-based for children and fixed for adults, with a gradual titration schedule to improve tolerability.
- Pediatric Patients (1 year of age and older):
- Initial Dose: Treatment should begin at a daily dose of 10 mg/kg, administered in two divided doses.[5]
- Titration: The dose should be increased by increments of approximately 10 mg/kg every other day until the target dose is reached.[22]
- Target and Maximum Dose: The recommended target maintenance dose is 45 mg/kg/day. The total daily dose should not exceed 3200 mg.[5]
- Adults (17 years of age and older):
- Initial Dose: Treatment should begin at a daily dose of 400 mg to 800 mg, administered in two divided doses.[5]
- Titration: The dose should be increased by 400 mg to 800 mg every other day.[5]
- Maximum Dose: The maximum recommended daily dose is 3200 mg, administered in two divided doses.[5]
- Dose Adjustment for Concomitant Valproate: A critical adjustment is required for patients who are also taking valproic acid. Because valproate inhibits the metabolism of Rufinamide, it can lead to significantly higher plasma concentrations. Therefore, in patients receiving valproate, Rufinamide should be initiated at a lower dose, and the titration should be more cautious. The maximum target dose may also need to be lower in these patients.[2]
6.4 Dosing in Special Populations
- Hepatic Impairment: Caution and careful dose titration are advised for patients with mild-to-moderate hepatic impairment. Rufinamide is not recommended for use in patients with severe hepatic impairment due to a lack of data.[2]
- Renal Impairment: The pharmacokinetics of Rufinamide are not significantly altered even in patients with severe renal failure. Therefore, no dose adjustment is generally required for patients with renal impairment.[2]
Table 3: Dosing and Titration Schedule for Rufinamide in LGS
| Patient Population | Initial Daily Dose | Titration Schedule | Maximum Recommended Daily Dose | Special Considerations |
|---|
| Pediatric Patients (≥1 year) | 10 mg/kg/day (in 2 divided doses) | Increase by 10 mg/kg every 2 days | 45 mg/kg/day, not to exceed 3200 mg/day | Dosing is weight-based. |
| Adults | 400–800 mg/day (in 2 divided doses) | Increase by 400–800 mg every 2 days | 3200 mg/day | Dosing is fixed. |
| Patients on Concomitant Valproate | A lower starting dose is recommended. | Titrate cautiously. | A lower maximum dose may be required. | Valproate significantly increases Rufinamide plasma concentrations. |
7.0 Safety and Tolerability Profile
The safety profile of Rufinamide has been well-characterized through clinical trials and post-marketing surveillance. While generally manageable, it includes several important risks that require careful patient selection and monitoring, including a unique cardiac contraindication and warnings common to the antiepileptic drug class.
7.1 Contraindications
There are two absolute contraindications for the use of Rufinamide:
- Familial Short QT Syndrome: Rufinamide is strictly contraindicated in patients with this rare, inherited cardiac channelopathy.[6] This is because Rufinamide has been shown to cause a dose-dependent shortening of the QT interval on the electrocardiogram (ECG). In individuals with Familial Short QT syndrome, who already have a pathologically short QT interval, further shortening can precipitate life-threatening ventricular arrhythmias, such as ventricular fibrillation, and increase the risk of sudden cardiac death.[6]
- Hypersensitivity: The drug is contraindicated in patients with a known hypersensitivity to the active substance, Rufinamide, to other triazole derivatives, or to any of the inactive ingredients in the formulation.[17]
7.2 Warnings and Precautions
Several significant warnings and precautions are associated with Rufinamide therapy.
- Suicidal Behavior and Ideation: In line with a class-wide warning for all antiepileptic drugs, Rufinamide increases the risk of suicidal thoughts or behavior. Pooled analyses of AED trials have shown that patients taking these drugs have approximately twice the risk compared to placebo.[6] Patients, their families, and caregivers must be informed of this risk and advised to immediately report any emergence or worsening of depression, unusual changes in mood or behavior, thoughts of self-harm, or suicidal ideation to their healthcare provider.[8]
- Central Nervous System (CNS) Reactions: The use of Rufinamide is frequently associated with CNS-related adverse reactions. These include somnolence (sleepiness) or fatigue, dizziness, ataxia (impaired coordination), and gait disturbances.[6] Patients should be cautioned about the potential for these effects to impair their ability to perform tasks requiring mental alertness, such as driving or operating heavy machinery, until they are familiar with how the medication affects them.[8]
- QT Interval Shortening: As noted above, Rufinamide shortens the cardiac QT interval. Formal ECG studies have demonstrated a mean reduction of up to 20 msec at higher doses.[6] While reductions below 300 msec have not been observed and the clinical risk in the general population is considered low, caution should be exercised when Rufinamide is co-administered with other medications that are also known to shorten the QT interval.[8] The unique nature of this cardiac risk, contrasting with the more common concern of QT
prolongation with other drugs, mandates careful consideration. It implies that a baseline ECG and a thorough family history for sudden cardiac death may be prudent before initiating therapy, especially if there is any clinical suspicion of an underlying cardiac condition.[11]
- Multi-organ Hypersensitivity / DRESS: Serious, potentially life-threatening systemic reactions have been reported with Rufinamide, including Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS), also known as multi-organ hypersensitivity.[6] These reactions typically present with fever, rash, and evidence of organ involvement (e.g., hepatitis, nephritis, lymphadenopathy). Clinical trial data suggest a specific window of vulnerability, with these reactions occurring primarily in children under 12 years of age and within the first four weeks of treatment.[6] This finding requires heightened clinical vigilance during the initiation phase of therapy in pediatric patients. Any rash, particularly if accompanied by systemic symptoms, must be considered a potential sign of a serious reaction, and Rufinamide should be discontinued immediately pending further evaluation.[8]
- Withdrawal of Therapy: As with all AEDs, Rufinamide should not be discontinued abruptly. Sudden withdrawal can precipitate an increase in seizure frequency, seizure exacerbation, or status epilepticus.[6] To minimize this risk, the dose should be tapered gradually, for instance, by reducing the total daily dose by approximately 25% every two days.[6]
- Status Epilepticus: In controlled clinical trials for LGS, a numerically higher incidence of episodes described as status epilepticus was observed in patients treated with Rufinamide (4.1%) compared to placebo (0%).[6] Clinicians should be aware of this potential risk.
7.3 Adverse Drug Reactions
The most frequently reported adverse effects of Rufinamide are CNS-related.
- Most Common Adverse Reactions (≥10% and more frequent than placebo): The most common side effects observed in pooled clinical trials were headache, dizziness, fatigue, somnolence, and nausea.[1]
- Adverse Reactions in Pediatric Patients: In children, somnolence and vomiting are particularly common, each occurring in 17% of patients in controlled trials. Other frequent adverse effects include headache, fatigue, dizziness, decreased appetite, rash, and ataxia.[6]
- Adverse Reactions in Adult Patients: In adults, headache and dizziness are the most frequent side effects. Other common reactions include fatigue, nausea, somnolence, diplopia (double vision), tremor, nystagmus, and blurred vision.[6]
7.4 Use in Specific Populations
- Pregnancy and Lactation: There are insufficient data from human pregnancies to assess the risk of Rufinamide. It should be used during pregnancy only if the potential benefit to the mother justifies the potential risk to the fetus.[2] Women of childbearing potential must be advised to use effective contraceptive measures during treatment. Pregnant patients are encouraged to enroll in the North American Antiepileptic Drug (NAAED) Pregnancy Registry to help collect safety data.[8] It is unknown whether Rufinamide is excreted in human breast milk, and breastfeeding should be avoided during treatment due to the potential for adverse effects in the infant.[2]
8.0 Drug-Drug Interactions
The drug-drug interaction (DDI) profile of Rufinamide is distinct, primarily governed by its non-CYP450 metabolism and its weak effects on the CYP3A4 enzyme. This creates a dual profile where Rufinamide is both a potential "victim" of interactions that affect its clearance and a "perpetrator" that can alter the metabolism of other drugs.
8.1 Rufinamide as a Victim: Effects of Other Drugs on Rufinamide
The plasma concentration and clearance of Rufinamide can be significantly altered by co-administered drugs that affect its primary metabolic pathway, carboxylesterase-mediated hydrolysis.
- Valproic Acid: This is the most clinically significant interaction affecting Rufinamide. Valproic acid is an inhibitor of carboxylesterase enzymes. When co-administered, it significantly decreases the clearance of Rufinamide, leading to a substantial increase in its plasma concentrations.[2] This interaction can increase the risk of dose-related adverse effects and necessitates a lower starting dose and more cautious titration of Rufinamide in patients taking valproate.[1]
- Potent Enzyme-Inducing AEDs: Broad-spectrum enzyme inducers, including carbamazepine, phenytoin, phenobarbital, and primidone, can increase the clearance of Rufinamide. While Rufinamide is not a CYP substrate, these drugs may induce the activity of carboxylesterases or other pathways involved in its metabolism and disposition. This interaction can lead to a decrease in Rufinamide plasma concentrations by 25% to 46%, potentially reducing its efficacy.[1]
8.2 Rufinamide as a Perpetrator: Effects of Rufinamide on Other Drugs
Rufinamide can alter the concentrations of other drugs, primarily through its weak induction of the CYP3A4 enzyme.
- CYP3A4 Substrates (including Hormonal Contraceptives): Rufinamide is a weak inducer of CYP3A4.[1] This action can accelerate the metabolism and decrease the plasma concentrations of drugs that are substrates of this enzyme. The most critical clinical consequence of this is the reduced efficacy of hormonal contraceptives (e.g., birth control pills containing ethinylestradiol and progestins), which are extensively metabolized by CYP3A4.[1] Co-administration of Rufinamide has been shown to decrease exposure to oral contraceptives, which may lead to contraceptive failure.[2] Therefore, women of childbearing potential using hormonal contraceptives are strongly advised to use an additional, effective, non-hormonal method of contraception during and for a period after treatment with Rufinamide.[2]
- Other Antiepileptic Drugs: The effects of Rufinamide on other AEDs are generally modest. Population pharmacokinetic modeling studies suggest that Rufinamide may slightly increase the clearance of carbamazepine and lamotrigine and slightly decrease the clearance of phenobarbital and phenytoin. The predicted changes are typically less than 20% and may not always be clinically significant, though it is recommended to consider a dose reduction of phenytoin when co-administered with Rufinamide.[29]
- CYP2E1 Substrates: As a weak inhibitor of CYP2E1, Rufinamide has the potential to increase the plasma levels of drugs metabolized by this enzyme, such as the muscle relaxant chlorzoxazone. However, the clinical significance of this in vitro finding has not been confirmed in dedicated clinical studies.[1]
- Central Nervous System Depressants: Rufinamide can cause CNS depression (e.g., somnolence, dizziness). When taken with other CNS depressants, including alcohol, benzodiazepines, or opioids, there is a risk of additive sedative and motor-impairing effects. Patients should be counseled on this potential for enhanced CNS depression.[1]
The DDI profile of Rufinamide requires a nuanced clinical approach. The initial perception of a "clean" profile due to its non-CYP450 metabolism is an oversimplification. Clinicians must recognize its dual nature: it is a victim of the profound inhibitory effect of valproate on its specific metabolic pathway, and it is a perpetrator through its weak but clinically meaningful induction of CYP3A4, which compromises the efficacy of hormonal contraceptives. Safe prescribing, especially in the polypharmacy environment of LGS, depends on a clear understanding of this duality.
Table 4: Clinically Significant Drug-Drug Interactions with Rufinamide
| Interacting Drug/Class | Effect on Rufinamide | Effect of Rufinamide on Other Drug | Mechanism | Clinical Recommendation |
|---|
| Valproic Acid | Plasma concentrations significantly increased | Minimal | Inhibition of carboxylesterase enzymes responsible for Rufinamide metabolism | Initiate Rufinamide at a lower dose and titrate more cautiously. A lower maximum dose may be necessary. |
| Enzyme-Inducing AEDs (Carbamazepine, Phenytoin, Phenobarbital) | Plasma concentrations decreased (by 25-46%) | Modest effects on some AEDs | Induction of Rufinamide metabolism (e.g., carboxylesterases) | Monitor for reduced Rufinamide efficacy; dose increase may be needed. |
| Hormonal Contraceptives | None | Plasma concentrations of contraceptive hormones decreased; potential for contraceptive failure | Weak induction of CYP3A4, which metabolizes contraceptive steroids | Advise patient to use an additional, effective, non-hormonal method of contraception. |
| Phenytoin | Plasma concentrations decreased | Plasma concentrations of Phenytoin may be slightly increased | Rufinamide may slightly decrease Phenytoin clearance | Monitor Phenytoin levels and consider a dose reduction of Phenytoin. |
| CNS Depressants (Alcohol, Benzodiazepines) | None | Additive CNS depressant effects (somnolence, dizziness, ataxia) | Pharmacodynamic interaction | Counsel patient on the risk of increased sedation and motor impairment. |
9.0 Regulatory Status and Development History
The development and approval of Rufinamide provide a clear example of a successful regulatory pathway for an orphan drug designed to treat a rare and severe disease. Its journey from initial development to global availability was facilitated by specific regulatory designations and a targeted clinical trial program.
9.1 Development and Manufacturer
Rufinamide, a novel triazole derivative, was initially developed by Novartis Pharma, AG, in 2004.[3] Subsequently, the rights were acquired by Eisai, a Japanese pharmaceutical company, which now manufactures and markets the drug globally.[3]
9.2 Orphan Drug Designation
Recognizing the significant unmet medical need in the LGS population, regulatory agencies in both the United States and Europe granted Rufinamide orphan drug status early in its development. This designation is reserved for drugs intended to treat rare diseases and provides incentives, such as market exclusivity and financial support, to encourage development.
- United States (FDA): Rufinamide was granted orphan designation by the FDA on October 8, 2004, for the adjunctive treatment of seizures associated with LGS.[10]
- European Union (EMA): The European Commission granted orphan designation for the same indication on October 20, 2004.[5] This status conferred a period of market exclusivity, which expired in January 2019.[37]
This early designation was instrumental in de-risking the development process and ensuring that the drug could be brought to a patient population with limited therapeutic options. It represents a successful application of regulatory frameworks designed to stimulate research and development for rare disorders.
9.3 Marketing Authorization Timeline
Rufinamide's approval process followed a logical progression, with initial authorization in Europe followed by the United States, and subsequent label extensions to include younger pediatric patients.
- European Union (EMA): Rufinamide was first authorized under the brand name Inovelon® on January 16, 2007. The initial approval was for adjunctive therapy in LGS for patients aged 4 years and older.[10] In August 2018, the European Commission extended this approval to include patients as young as one year of age.[34]
- United States (FDA): The FDA first approved Rufinamide under the brand name Banzel® on November 14, 2008. The indication was for the adjunctive treatment of seizures associated with LGS in children aged 4 years and older and in adults.[11]
- On March 4, 2011, the FDA approved the 40 mg/mL oral suspension formulation, enhancing its utility in the pediatric population.[36]
- On February 13, 2015, the FDA extended the indication to include pediatric patients from one to four years of age.[33]
9.4 Pediatric Label Extension
The extension of the approved age range from ≥4 years down to ≥1 year is a noteworthy aspect of Rufinamide's regulatory history. This age de-escalation was not based on a new, large-scale, placebo-controlled efficacy trial in the younger population, which would be ethically and logistically challenging to conduct. Instead, regulators accepted a modern, data-driven approach using a pharmacokinetic (PK) bridging study (Study 303).[30] This study demonstrated that the pharmacokinetic profile of Rufinamide in children aged 1 to <4 years was similar to that observed in older children and adults, and that the safety profile was consistent.[30] By showing that similar drug exposures could be achieved safely in the younger population, efficacy could be reasonably extrapolated from the pivotal trial data in older patients. This represents an efficient and ethical regulatory strategy that facilitates timely access to necessary medications for vulnerable pediatric populations.
9.5 Generic Availability
Following the expiration of patents and market exclusivity periods, generic versions of Rufinamide have become available, increasing access and potentially reducing healthcare costs.[3]
10.0 Synthesis and Concluding Remarks
Rufinamide has established itself as a significant and valuable agent in the armamentarium for treating Lennox-Gastaut Syndrome. Its journey from a structurally novel compound to a globally approved therapy is a testament to targeted drug development for rare and complex neurological disorders. This monograph provides a comprehensive synthesis of its chemical, pharmacological, and clinical characteristics.
10.1 Consolidated Profile
Rufinamide is a triazole-derivative antiepileptic drug, a structural class that sets it apart from all other AEDs. Its primary mechanism of action, the stabilization of the inactive state of voltage-gated sodium channels, is complemented by evidence of secondary actions on glutamate receptors and potassium channels, suggesting a multi-modal approach to dampening neuronal hyperexcitability. This complex pharmacology may be key to its notable success in the challenging, multi-faceted seizure environment of LGS.
Its pharmacokinetic profile is defined by a unique, non-cytochrome P450 metabolic pathway mediated by carboxylesterases. This feature provides a distinct advantage by avoiding many common drug-drug interactions. However, this profile is not without its own specific challenges, namely a profound interaction with the carboxylesterase inhibitor valproic acid and a clinically relevant induction of CYP3A4, which impacts the efficacy of hormonal contraceptives. These interactions demand a high level of clinical vigilance and patient counseling.
10.2 Place in Therapy
The therapeutic niche for Rufinamide is clearly and firmly established as an adjunctive therapy for LGS. Its clinical value is anchored by robust, placebo-controlled evidence demonstrating a significant reduction in seizure frequency, with a particularly powerful and clinically meaningful impact on the tonic-atonic "drop attack" seizures that are a major source of injury and disability in this population. The availability of patient-friendly formulations, such as a bioequivalent oral suspension and crushable tablets, further solidifies its utility in a patient group that often includes young children and individuals with swallowing difficulties. Its safety profile, while requiring careful management of CNS side effects and vigilance for rare but serious hypersensitivity reactions, is generally considered manageable. The absolute contraindication in Familial Short QT syndrome remains a critical, albeit rare, consideration that necessitates careful patient screening.
10.3 Future Research Directions
While Rufinamide's role in LGS is well-defined, several avenues for future research could further clarify and potentially expand its utility:
- Mechanistic Elucidation: Further preclinical and clinical research is warranted to fully elucidate the contribution of its secondary mechanisms of action (mGluR5 inhibition, BKCa channel stimulation) to its overall clinical effect. Understanding how these pathways interact could inform the development of next-generation AEDs with even greater efficacy.
- Exploration of Other Indications: The mechanistic rationale for its use in other conditions characterized by neuronal hyperexcitability remains compelling. Despite an earlier withdrawn trial, well-designed studies in specific neuropathic pain syndromes could be considered. Similarly, its potential in other severe epileptic encephalopathies beyond LGS warrants further investigation.
- Comparative Effectiveness Research: The LGS treatment landscape has evolved since Rufinamide's approval, with the introduction of other effective agents like clobazam and cannabidiol. Head-to-head comparative effectiveness trials are needed to better define the optimal sequencing and combination of these therapies, helping clinicians to create more evidence-based, individualized treatment algorithms for patients with LGS.
10.4 Final Conclusion
In conclusion, Rufinamide represents a significant therapeutic advance for patients with Lennox-Gastaut Syndrome. It offers a unique combination of a novel chemical structure, a multi-modal mechanism of action, targeted clinical efficacy against the most debilitating seizures, and a distinct pharmacokinetic profile. While its use requires careful attention to specific safety considerations and drug interactions, it has rightfully earned its place as a cornerstone of therapy for this severe and challenging epileptic encephalopathy. Its development and successful regulatory approval serve as a powerful illustration of how targeted pharmaceutical innovation, supported by dedicated regulatory pathways like the orphan drug program, can deliver meaningful clinical benefits to underserved patient populations.
Works cited
- Rufinamide: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed September 13, 2025, https://go.drugbank.com/drugs/DB06201
- Rufinamide | 106308-44-5 - ChemicalBook, accessed September 13, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB5972298.htm
- Rufinamide - Wikipedia, accessed September 13, 2025, https://en.wikipedia.org/wiki/Rufinamide
- Rufinamide: Uses, Dosage, Side Effects, Warnings - Drugs.com, accessed September 13, 2025, https://www.drugs.com/rufinamide.html
- Rufinamide in patients with Lennox-Gastaut syndrome | NRR - Dove Medical Press, accessed September 13, 2025, https://www.dovepress.com/rufinamide-in-patients-with-lennox-gastaut-syndrome-peer-reviewed-fulltext-article-NRR
- BANZEL™ (rufinamide) Tablets DESCRIPTION BANZEL (rufinamide ..., accessed September 13, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/021911lbl.pdf
- Australian Public Assessment Report for Rufinamide - Therapeutic Goods Administration (TGA), accessed September 13, 2025, https://www.tga.gov.au/sites/default/files/auspar-rufinamide-190321.pdf
- BANZEL (rufinamide) | BANZEL.com, accessed September 13, 2025, https://www.banzel.com/
- Role of rufinamide in the management of Lennox-Gastaut syndrome (childhood epileptic encephalopathy) - PMC - PubMed Central, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2654531/
- Efficacy and safety of rufinamide in pediatric epilepsy - PMC, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3625016/
- Rufinamide - Epilepsy Foundation, accessed September 13, 2025, https://www.epilepsy.com/tools-resources/seizure-medication-list/rufinamide
- Rufinamide (CGP 33101, RUF 331, CAS Number: 106308-44-5) | Cayman Chemical, accessed September 13, 2025, https://www.caymanchem.com/product/18870/rufinamide
- Rufinamide | CAS 106308-44-5 | SCBT - Santa Cruz Biotechnology, accessed September 13, 2025, https://www.scbt.com/p/rufinamide-106308-44-5
- Rufinamide | CAS 106308-44-5 - LGC Standards, accessed September 13, 2025, https://www.lgcstandards.com/US/en/Rufinamide/p/TRC-R701550?queryID=4c799054d39dd0397b70c39d0a20583c
- en.wikipedia.org, accessed September 13, 2025, https://en.wikipedia.org/wiki/Rufinamide#:~:text=However%2C%20several%20recent%20clinical%20trials,available%20as%20a%20generic%20medication.
- Rufinamide Tablets - Cleveland Clinic, accessed September 13, 2025, https://my.clevelandclinic.org/health/drugs/20188-rufinamide-tablets
- Inovelon | European Medicines Agency (EMA), accessed September 13, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/inovelon
- Rufinamide | C10H8F2N4O - ChemSpider, accessed September 13, 2025, https://www.chemspider.com/Chemical-Structure.114471.html
- Rufinamide | C10H8F2N4O | CID 129228 - PubChem, accessed September 13, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Rufinamide
- rufinamide | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 13, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=7470
- CAS No : 106308-44-5| Product Name : Rufinamide - API - Pharmaffiliates, accessed September 13, 2025, https://www.pharmaffiliates.com/en/106308-44-5-rufinamide-api-pa1862000.html
- rufinamide tablets - accessdata.fda.gov, accessed September 13, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/204988Orig1s000lbl.pdf
- Rufinamide - StatPearls - NCBI Bookshelf, accessed September 13, 2025, https://www.ncbi.nlm.nih.gov/books/NBK557595/
- Rufinamide Improves Functional and Behavioral Deficits via Blockade of Tetrodotoxin-Resistant Sodium Channels in Diabetic Neuropathy - PubMed, accessed September 13, 2025, https://pubmed.ncbi.nlm.nih.gov/26036975/
- Rufinamide | Na+ channel Blocker - Hello Bio, accessed September 13, 2025, https://hellobio.com/rufinamide.html
- Rufinamide, a Triazole-Derived Antiepileptic Drug, Stimulates Ca2+-Activated K+ Currents While Inhibiting Voltage-Gated Na+ Currents - PMC - PubMed Central, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9697614/
- Rufinamide Withdrawn Phase 2 Trials for Pain / Peripheral Nerve Injury (PNI) Treatment, accessed September 13, 2025, https://go.drugbank.com/drugs/DB06201/clinical_trials?conditions=DBCOND0012160%2CDBCOND0059871&phase=2&purpose=treatment&status=withdrawn
- Mechanism of Action | BANZEL (rufinamide), accessed September 13, 2025, https://www.banzel.com/hcp/mechanism-of-action
- Rufinamide: Clinical Pharmacokinetics and concentration-response relationships in patients with epilepsy | Request PDF - ResearchGate, accessed September 13, 2025, https://www.researchgate.net/publication/5345854_Rufinamide_Clinical_Pharmacokinetics_and_concentration-response_relationships_in_patients_with_epilepsy
- Pivotal Trial Design | BANZEL (rufinamide), accessed September 13, 2025, https://www.banzel.com/hcp/study-design
- Rufinamide from clinical trials to clinical practice in the United States and Europe - PubMed, accessed September 13, 2025, https://pubmed.ncbi.nlm.nih.gov/21669560/
- Rufinamide Completed Phase 2 Trials for Generalized Anxiety Disorder Treatment, accessed September 13, 2025, https://go.drugbank.com/drugs/DB06201/clinical_trials?conditions=DBCOND0027966&phase=2&purpose=treatment&status=completed
- Eisai Announces FDA Approval of BANZEL® (rufinamide) for Adjunctive Treatment of Seizures Associated with Lennox-Gastaut Syndrome in Pediatric Patients Ages 1-4 - Feb 13, 2015, accessed September 13, 2025, https://media-us.eisai.com/2015-02-13-Eisai-Announces-FDA-Approval-of-BANZEL-rufinamide-for-Adjunctive-Treatment-of-Seizures-Associated-with-Lennox-Gastaut-Syndrome-in-Pediatric-Patients-Ages-1-4
- Eisai Receives Marketing Authorisation Extension for Inovelon from the European Commission - FirstWord Pharma, accessed September 13, 2025, https://firstwordpharma.com/story/4643663
- Final Population Pharmacokinetic Parameter Estimates of Rufinamide - ResearchGate, accessed September 13, 2025, https://www.researchgate.net/figure/Final-Population-Pharmacokinetic-Parameter-Estimates-of-Rufinamide_tbl2_290222019
- Banzel (rufinamide) FDA Approval History - Drugs.com, accessed September 13, 2025, https://www.drugs.com/history/banzel.html
- EU/3/04/240 - orphan designation for treatment of Lennox-Gastaut syndrome, accessed September 13, 2025, https://www.ema.europa.eu/en/medicines/human/orphan-designations/eu-3-04-240
- Drug Approval Package: Banzel (Rufinamide) NDA #021911 - accessdata.fda.gov, accessed September 13, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2008/021911s000_TOC.cfm
- 201367 Rufinamide Clinical BPCA | FDA, accessed September 13, 2025, https://www.fda.gov/files/drugs/published/201367-Rufinamide-Clinical-BPCA.pdf