Zonegran, Zonisade, Zonisamide Mylan, Zonisamide Viatris (previously Zonisamide Mylan)
Small Molecule
C8H8N2O3S
68291-97-4
Partial-Onset Seizures
Zonisamide is a second-generation, broad-spectrum antiepileptic drug (AED) distinguished by its unique 1,2-benzisoxazole chemical structure and its classification as a sulfonamide.[1] Its therapeutic efficacy is attributed to a multi-modal mechanism of action that is not yet fully elucidated but is primarily understood to involve the blockade of voltage-gated sodium channels and low-threshold (T-type) calcium channels. These primary actions are complemented by secondary modulatory effects on major neurotransmitter systems, including the GABAergic, glutamatergic, dopaminergic, and serotonergic pathways.[1]
The primary approved indication for zonisamide in the United States and Europe is as an adjunctive therapy for the treatment of partial-onset seizures in adults and, in some jurisdictions, older pediatric patients.[1] The drug's pharmacokinetic profile is notable for its high oral bioavailability, exceptionally long elimination half-life, and extensive, saturable binding to erythrocytes, which acts as a significant drug reservoir and necessitates a prolonged period to achieve steady-state concentrations.[2]
The clinical use of zonisamide must be carefully balanced against its significant safety considerations, which are mechanistically linked to its fundamental chemical properties. As a sulfonamide, it carries a risk of severe and potentially fatal hypersensitivity reactions, including Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN). Furthermore, its weak but clinically relevant activity as a carbonic anhydrase inhibitor predisposes patients to metabolic acidosis and nephrolithiasis, requiring diligent monitoring.[11] This monograph provides an exhaustive review of zonisamide's chemistry, pharmacology, clinical efficacy, and safety profile to guide its optimal use in clinical practice.
The clinical availability and therapeutic application of zonisamide have been shaped by a protracted and fragmented developmental history spanning several decades and multiple corporate entities. This complex journey from its initial discovery in Japan to its eventual, staggered approval in Western markets has resulted in significant regional disparities in its approved indications and has influenced its perception within the global medical community.
Zonisamide was first synthesized by Uno and colleagues at Dainippon Pharmaceutical (now Dainippon Sumitomo Pharma) in Japan in 1972.[13] The initial line of inquiry for the compound was not for epilepsy but for the treatment of psychiatric diseases.[6] However, subsequent preclinical screening revealed its potent anticonvulsant properties. In the maximal electroshock (MES) test, a standard preclinical model for identifying agents that prevent seizure spread and are effective against generalized tonic-clonic seizures, zonisamide demonstrated efficacy comparable to that of established AEDs like phenobarbital and carbamazepine.[14] This early evidence established its potential as a broad-spectrum anticonvulsant. Following successful clinical development, Dainippon launched the drug in Japan in 1989 under the brand name Excegran for the treatment of epilepsy.[1] It gained approval in Korea shortly thereafter in 1990.[18]
The introduction of zonisamide into Western markets was considerably more complicated and delayed. Initial development in the United States was undertaken by Warner-Lambert/Parke-Davis, which, for reasons not fully detailed in the public record, discontinued its program in 1987.[18] Dainippon then re-assumed responsibility for development in the US, conducting further studies until 1997. At that point, the rights to zonisamide were acquired by Athena Neurosciences, a company that would later become part of Elan Pharmaceuticals.[18]
The path to European approval was similarly fraught. A Marketing Authorisation Application submitted to European regulators in 1997 was ultimately withdrawn. The Committee for Proprietary Medicinal Products (CPMP) identified major deficiencies in the dossier, notably that the pivotal clinical trials submitted were of insufficient duration to adequately assess long-term efficacy, reflecting the increasingly stringent regulatory standards in Europe.[18]
After this setback, Elan Pharmaceuticals successfully navigated the US regulatory process, securing Food and Drug Administration (FDA) approval for Zonegran (zonisamide) on March 1, 2000. The approved indication was for adjunctive therapy in the treatment of partial-onset seizures in adults.[1] European approval did not follow until 2005, a full 16 years after its Japanese debut.[1] In 2004, Elan transferred its interests in zonisamide to Eisai Co., Ltd., which has since managed the marketing of the drug in the United States, Europe, and various countries in Asia.[13]
The long and separate development timelines in Japan versus the West fostered an environment where clinical experience and research priorities diverged. This divergence is most clearly illustrated by the approval of zonisamide for the treatment of Parkinson's disease (PD) in Japan. This indication, granted in 2009 as an adjunctive therapy to levodopa, reportedly stemmed from a serendipitous clinical observation in a patient who had both epilepsy and PD.[13] With nearly a decade of post-marketing experience before the drug was even approved in the US, Japanese clinicians and the local developer, Dainippon, were uniquely positioned to identify and formally investigate this novel therapeutic effect. In contrast, Western development, managed by different companies and facing stricter regulatory hurdles, remained narrowly focused on establishing efficacy in epilepsy to meet the demands of the FDA and the European Medicines Agency (EMA). Consequently, the use of zonisamide for PD remains an off-label or investigational application in most of the world.[21]
A more recent significant development occurred in July 2022, when the FDA approved ZONISADE™, the first oral suspension formulation of zonisamide.[22] Developed by Eton Pharmaceuticals and commercialized by Azurity Pharmaceuticals, this new formulation was designed to address the long-standing clinical challenge of administering the drug to patients with dysphagia, including certain pediatric and geriatric populations, who are unable to swallow the solid capsule form.[23]
The therapeutic application and pharmaceutical development of zonisamide are fundamentally governed by its distinct chemical structure and physical properties. These characteristics have dictated its formulation, influenced its stability, and provided the basis for its classification as a unique antiepileptic agent.
Zonisamide is a synthetic small molecule identified chemically as 1,2-benzisoxazole-3-methanesulfonamide.[1] It is classified as a sulfonamide due to the presence of a sulfamoyl group (
−SO2NH2) but is structurally unrelated to other sulfonamide-containing AEDs or to other classes of antiepileptic agents.[1] The core of the molecule is a 1,2-benzisoxazole ring system. Zonisamide does not contain any chiral centers or optically active centers and therefore does not exhibit optical isomerism.[18]
Its molecular formula is C8H8N2O3S, corresponding to a molecular weight of 212.23 g/mol.[1] The compound is registered under the Chemical Abstracts Service (CAS) Number 68291-97-4 and has been assigned the DrugBank accession number DB00909.[1] A consolidated table of its key chemical and physical properties is provided below (Table 1).
Zonisamide exists as a white to pale yellow crystalline powder or as white needles.[1] It is reported to be both odorless and tasteless.[1] The compound is very slightly or moderately soluble in water, with a reported solubility of approximately 0.80 mg/mL.[1] Its solubility in 0.1 N HCl is slightly lower, at 0.50 mg/mL.[16] It is more readily soluble in organic solvents such as methanol, ethanol, and ethyl acetate.[1]
The melting point is consistently reported in the range of 161–165 °C [1], although some sources indicate decomposition occurs at a much higher temperature of 275 °C.[2] Zonisamide is a weak acid with a dissociation constant (
pKa) of 10.2.[1] It is described as a very stable and non-hygroscopic compound, which simplifies its storage and handling.[18]
The inherent physicochemical properties of zonisamide have been the primary determinants of its pharmaceutical formulation. The combination of poor water solubility and a bitter taste presented a challenge for developing a palatable and bioavailable oral dosage form. This challenge directly led to the development of hard gelatin capsules as the initial and, for many years, the only available formulation. This approach effectively masked the taste and, through optimization of drug particle size and the use of suitable excipients, achieved adequate dissolution and bioavailability.[18]
For over two decades following its US approval, this capsule-only availability created a significant clinical gap for patient populations with difficulty swallowing, such as young children, the elderly, and those with neurological or physical impairments causing dysphagia. This long-standing unmet need was finally addressed in 2022 with the introduction of a liquid formulation.
The currently available formulations are:
Table 1: Physical and Chemical Properties of Zonisamide
Property | Value / Identifier | Source(s) |
---|---|---|
IUPAC Name | 1,2-benzoxazol-3-ylmethanesulfonamide | 1 |
Chemical Formula | C8H8N2O3S | 1 |
Molecular Weight | 212.23 g/mol | 1 |
CAS Number | 68291-97-4 | 1 |
DrugBank ID | DB00909 | 1 |
InChIKey | UBQNRHZMVUUOMG-UHFFFAOYSA-N | 1 |
SMILES | C1=CC=C2C(=C1)C(=NO2)CS(=O)(=O)N | 1 |
Physical State | Crystalline solid / powder | 14 |
Color | White to pale yellow | 1 |
Melting Point | 161-165 °C | 1 |
pKa | 10.2 | 1 |
Water Solubility | 0.80 mg/mL | 16 |
LogP | 0.5 | 1 |
Zonisamide's clinical utility is derived from a complex and multi-modal pharmacological profile. While its precise mechanism of action remains to be fully elucidated, its anticonvulsant effects are believed to result from a combination of actions on voltage-gated ion channels, modulation of several major neurotransmitter systems, and weak enzymatic inhibition. This multifaceted activity may explain its broad-spectrum efficacy, particularly in patients with epilepsy refractory to other agents.[5] Its unique pharmacokinetic properties, especially its distribution and long half-life, are critical determinants of its clinical use.
Zonisamide exerts its therapeutic effects through several distinct but complementary mechanisms.
The principal antiepileptic actions of zonisamide are attributed to its effects on neuronal ion channels.
Zonisamide also influences several key neurotransmitter systems, although the clinical significance of these actions is less defined than its effects on ion channels.
Zonisamide is a weak inhibitor of the enzyme carbonic anhydrase, with activity against several human isoforms, including CA-I, CA-II, and CA-V.[1] While this property was initially thought to be a primary mechanism, similar to the sulfonamide AED acetazolamide, it is now widely accepted that this action is not a major contributor to zonisamide's primary
antiepileptic effects.[4] However, this weak enzymatic inhibition is clinically highly significant, as it is the direct cause of notable adverse effects, including the development of metabolic acidosis and an increased risk of renal stone formation.[4]
Evidence from preclinical studies suggests that zonisamide may possess neuroprotective properties that are independent of its direct anticonvulsant activity.[32] It has been shown to act as an antioxidant, scavenging free radicals and protecting against oxidative stress-induced neuronal injury.[1] This is supported by findings in animal models where zonisamide prevented the reduction of dopamine and its metabolites in the striatum following exposure to the neurotoxin MPTP.[14]
The composite pharmacodynamic effect of zonisamide's multi-modal actions is the suppression of abnormal neuronal excitability, leading to an increase in seizure threshold and a reduction in the initiation and propagation of seizure activity.[9] These same mechanisms also underlie its adverse effect profile. The direct extension of its primary CNS-modulating activity results in common side effects such as somnolence, dizziness, and cognitive slowing. The most severe and life-threatening adverse reactions, such as SJS/TEN, DRESS, and other blood dyscrasias, are idiosyncratic events linked to its sulfonamide chemical structure and the potential for severe hypersensitivity.[9]
The clinical use of zonisamide is heavily influenced by its unique pharmacokinetic profile, characterized by rapid absorption, extensive distribution into erythrocytes, and a very long elimination half-life.
Following oral administration, zonisamide is rapidly and completely absorbed, exhibiting high oral bioavailability of approximately 95%.[4] In healthy volunteers, peak plasma concentrations (
Tmax) are typically reached within 2 to 6 hours.[4] The presence of food delays the time to reach peak concentration but does not affect the overall extent of absorption (i.e., its bioavailability is unchanged).[4]
Zonisamide has an apparent volume of distribution (Vd/F) of approximately 1.45 L/kg.[9] Its distribution is defined by two key features:
This binding to erythrocytes is also saturable.[4] This saturation phenomenon explains why the drug's pharmacokinetics become non-linear at high doses (e.g., >800 mg), as the erythrocyte reservoir becomes full, causing a disproportionate increase in free plasma concentrations.[9] It also underlies early clinical observations that monitoring of whole
blood concentrations might provide a more stable indicator of drug exposure than monitoring serum or plasma concentrations alone, as the latter can show greater inter-patient variability.[10] This suggests that standard therapeutic drug monitoring of plasma levels may not fully capture the total body drug load, a critical consideration in managing complex or refractory patients.
Zonisamide undergoes extensive hepatic metabolism, primarily via reductive and conjugative pathways, with oxidation playing only a minor role.[4] The major metabolic pathway is the reductive cleavage of the benzisoxazole ring by cytochrome P450 3A4 (CYP3A4) to form the open-ring metabolite, 2–sulfamoylacetylphenol (SMAP).[4] CYP2C19 and CYP3A5 may also contribute to this process to a lesser extent.[4] A secondary metabolic pathway involves acetylation via N-acetyl-transferases to form N-acetyl zonisamide.[9]
A clinically important feature is that zonisamide does not induce its own metabolism (auto-induction) and does not significantly induce or inhibit other major hepatic enzymes. This characteristic minimizes its potential to alter the metabolism of co-administered drugs, which is a favorable attribute in polypharmacy settings common in epilepsy treatment.[4]
Zonisamide and its metabolites are eliminated primarily by the kidneys, with the majority of the dose recovered in the urine and only a small fraction in the feces.[9] Following administration of a radiolabeled dose, approximately 35% is excreted as unchanged zonisamide, 15% as N-acetyl zonisamide, and 50% as the glucuronide conjugate of SMAP.[9]
The elimination half-life of zonisamide is exceptionally long, averaging approximately 63 hours in plasma and an even longer 105 hours in red blood cells, due to the slow release from its erythrocyte reservoir.[2] This long half-life allows for convenient once- or twice-daily dosing regimens but also means that it takes approximately 14 days of consistent dosing to reach steady-state plasma concentrations.[2]
The plasma clearance of zonisamide is approximately 0.30–0.35 mL/min/kg in patients not receiving concomitant enzyme-inducing AEDs. This clearance is significantly increased (to approximately 0.5 mL/min/kg) in patients taking potent CYP3A4 inducers such as phenytoin, carbamazepine, or phenobarbital, which can necessitate dosage adjustments.[9]
Table 2: Key Pharmacokinetic Parameters of Zonisamide
Parameter | Value / Description | Source(s) |
---|---|---|
Oral Bioavailability | ~95% | 9 |
Time to Peak (Tmax) | 2–6 hours (fasted); delayed by food | 4 |
Plasma Protein Binding | ~40% | 4 |
Volume of Distribution (Vd/F) | ~1.45 L/kg | 9 |
Elimination Half-life | Plasma: ~63 hours; Erythrocytes: ~105 hours | 4 |
Time to Steady State | ~14 days | 4 |
Metabolism Pathway | Hepatic; Reduction to SMAP and Acetylation | 4 |
Primary Enzyme | Cytochrome P450 3A4 (CYP3A4) | 4 |
Route of Excretion | Primarily renal | 9 |
% Unchanged in Urine | ~35% | 9 |
Clearance (no inducers) | ~0.30–0.35 mL/min/kg | 9 |
Clearance (with inducers) | ~0.5 mL/min/kg | 9 |
The clinical utility of zonisamide has been most rigorously established in the treatment of epilepsy, its primary indication. However, its broad pharmacological profile and observed clinical effects have prompted significant investigation and off-label use in a variety of other neurological and psychiatric conditions. This pattern of application demonstrates a form of "reverse translation," where the drug's known secondary actions and side effect profile have directly informed its exploration in new, mechanistically plausible therapeutic areas.
In the United States, zonisamide is approved by the FDA as an adjunctive therapy for the treatment of partial-onset seizures in adults and pediatric patients aged 16 years and older.[1] Its efficacy for this indication was established in three pivotal multicenter, placebo-controlled, double-blind clinical trials conducted in patients with refractory epilepsy.[34]
These trials enrolled patients who were experiencing at least four partial-onset seizures per month despite receiving stable, therapeutic doses of one or two other AEDs.[34] Patients were randomized to receive either zonisamide or placebo as add-on therapy. The study protocols involved a dose-titration phase followed by a maintenance phase of at least 12 weeks.[18] The primary efficacy endpoint was the median percent reduction in partial seizure frequency from baseline, with the responder rate (the proportion of patients achieving a 50% or greater reduction in seizure frequency) serving as a key secondary endpoint.[34]
The results from these trials consistently demonstrated the statistically significant superiority of zonisamide over placebo. For instance, in a large US trial, patients receiving a maintenance dose of 400 mg/day of zonisamide experienced a median reduction in seizure frequency of 40.5%, compared to only a 9.0% reduction in the placebo group (p = 0.0009). The responder rate in this study was 42% for the zonisamide group.[34] The trials also established a clear dose-response relationship, with 100 mg/day identified as the minimal effective dose and 400 mg/day as the most effective dose studied.[37] Efficacy was found to be comparable whether the total daily dose was administered once daily or divided into two daily doses.[34]
Table 3: Summary of Pivotal Efficacy Trials of Zonisamide in Partial-Onset Seizures
Study Identifier / Reference | Design | Patient Population (n) | Treatment Arms (Dose) | Median % Seizure Reduction (Drug vs. Placebo) | Responder Rate (Drug vs. Placebo) |
---|---|---|---|---|---|
Study 1 34 | Randomized, Double-Blind, Placebo-Controlled, Dose-Response | 203 adults with refractory partial-onset seizures | Zonisamide 100, 200, 400 mg/day vs. Placebo | 40.5% (at 400 mg) vs. 9.0% | 41.8% (at 400 mg) vs. 22.2% |
Study 2 34 | Randomized, Double-Blind, Placebo-Controlled | 141 adults with refractory partial-onset seizures | Zonisamide (titrated) vs. Placebo | 29.6% vs. -3.2% | 29.0% vs. 15.0% |
Study 3 34 | Randomized, Double-Blind, Placebo-Controlled | 133 adults with refractory partial-onset seizures | Zonisamide (titrated) vs. Placebo | 27.2% vs. -1.1% | 28.0% vs. 12.0% |
The multifaceted pharmacology of zonisamide has led to its use and investigation in a wide range of conditions beyond its approved indication.
Zonisamide is frequently used off-label in the management of severe epilepsy syndromes, including infantile spasms (West syndrome), Lennox-Gastaut syndrome, and progressive myoclonic epilepsy.[2] Clinical trials have formally investigated its utility in other seizure types. A study of adjunctive zonisamide for primary generalized tonic-clonic seizures (NCT00692003) has been conducted.[39] Another trial in patients with myoclonus-dystonia (NCT01806805) demonstrated that zonisamide significantly improved action myoclonus and myoclonus-related functional disability compared to placebo.[40]
The exploration of zonisamide in PD is a direct consequence of its known effects on dopaminergic neurotransmission.[9] It is approved in Japan as an adjunct to levodopa for improving motor symptoms, typically at low doses of 25–50 mg per day.[13] Its mechanism in PD is thought to involve not only the facilitation of dopamine release but also potential inhibition of monoamine oxidase-B (MAO-B) and neuroprotective effects.[20]
The well-documented adverse effect of anorexia and weight loss has been repurposed as a potential therapeutic effect. Zonisamide is used off-label to treat binge-eating disorder, often in combination with cognitive behavioral therapy, where it is thought to act on appetite centers in the brain.[25] It has also been formally studied for
obesity, with trials showing significant positive effects on body weight loss, although a planned combination product (bupropion/zonisamide, brand name Empatic) had its development discontinued.[13]
Leveraging the same neuronal stabilizing properties that are effective in epilepsy, zonisamide is used off-label as a prophylactic treatment for migraine headaches. It is often considered as an alternative for patients who cannot tolerate or do not respond to topiramate, another AED used for migraine prevention.[2]
The CNS-modulating effects of zonisamide have prompted exploration in other areas. A Phase 2 clinical trial (NCT01765608) has been completed for the treatment of obstructive sleep apnea.[44] A Phase 1/2 trial (NCT01137890) has been completed to evaluate its effect on
cocaine addiction, likely exploring its impact on the brain's dopaminergic reward pathways.[45] Additionally, small open-label trials have suggested a potential benefit in treating
tardive dyskinesia, and it has been used off-label by psychiatrists as a mood stabilizer for bipolar depression.[2]
The clinical application of zonisamide requires a thorough understanding of its safety profile, which is characterized by a range of adverse effects from common, manageable CNS symptoms to rare, but severe and potentially fatal, systemic reactions. A sophisticated approach to risk management recognizes that these risks are not a random collection of events but fall into distinct clusters that are mechanistically linked to the drug's fundamental chemical structure and pharmacological actions.
The most common adverse drug reactions (ADRs) associated with zonisamide are predominantly related to its effects on the central nervous system. These include somnolence (drowsiness), dizziness, ataxia (impaired coordination), agitation or irritability, and cognitive difficulties such as problems with memory, concentration, or mental slowing. Anorexia (loss of appetite), which frequently leads to weight loss, is also a very common side effect.[2] A more comprehensive list of reported adverse effects, categorized by system organ class, includes gastrointestinal disturbances (nausea, diarrhea), psychiatric symptoms (depression, psychosis), and various other less frequent events.[25]
Zonisamide is contraindicated in patients with a known history of hypersensitivity to zonisamide or any other sulfonamide-containing drugs.[4] Beyond this absolute contraindication, several serious warnings and precautions demand careful patient selection and monitoring. These risks can be understood as three mechanistically-linked clusters.
As a sulfonamide derivative, zonisamide carries an inherent risk of inducing severe, idiosyncratic hypersensitivity reactions, which can be life-threatening. This risk is highlighted in a boxed warning on some international labels and is a critical consideration for prescribers.[4] These reactions include:
Zonisamide's weak inhibition of carbonic anhydrase is directly responsible for a distinct set of metabolic and renal adverse effects.
The therapeutic CNS-modulating activity of zonisamide can also lead to significant adverse effects.
Zonisamide is subject to several clinically significant interactions that can affect its efficacy and safety.
Zonisamide can be administered with or without food.[8] While food can delay the time it takes to reach peak plasma concentration, it does not impact the overall bioavailability of the drug.[4] Some sources advise caution with consuming large amounts of grapefruit juice, as it is a known inhibitor of intestinal CYP3A4 and could theoretically increase zonisamide absorption and the risk of adverse effects.[4]
Table 4: Clinically Significant Drug Interactions with Zonisamide
Interacting Drug/Class | Mechanism | Clinical Effect | Management Recommendation | Source(s) |
---|---|---|---|---|
CYP3A4 Inducers (e.g., Phenytoin, Carbamazepine, Phenobarbital, Rifampin) | Increased metabolic clearance of zonisamide via CYP3A4 induction. | Decreased zonisamide plasma concentration and half-life; potential loss of efficacy. | Monitor for efficacy. May require higher zonisamide maintenance dose and faster titration. | 9 |
CYP3A4 Inhibitors (e.g., Ketoconazole, Itraconazole, Clarithromycin) | Decreased metabolic clearance of zonisamide via CYP3A4 inhibition. | Increased zonisamide plasma concentration; increased risk of dose-related adverse effects. | Monitor for toxicity. May require lower zonisamide dose or slower titration. | 4 |
Other Carbonic Anhydrase Inhibitors (e.g., Topiramate, Acetazolamide) | Additive pharmacodynamic effect (inhibition of carbonic anhydrase). | Increased risk of metabolic acidosis and nephrolithiasis. | Use with caution. Monitor serum bicarbonate and for symptoms of kidney stones. | 9 |
CNS Depressants (e.g., Alcohol, Benzodiazepines, Opioids) | Additive pharmacodynamic effect (CNS depression). | Increased somnolence, dizziness, and cognitive/motor impairment. | Counsel patient to avoid or limit alcohol. Use with caution with other sedating drugs. | 50 |
Experience with acute zonisamide overdose is limited, but available case reports suggest a relatively benign clinical course with supportive care, even following massive ingestions of up to 12.6 g.[47] There is no specific antidote for zonisamide toxicity.
The clinical presentation of an overdose is an extension of the drug's known pharmacological effects. The most prominent features are related to CNS depression and may include profound somnolence, progressing to coma, as well as ataxia and vomiting.[46] Cardiovascular effects such as bradycardia and hypotension, as well as respiratory depression, have also been reported.[46] Due to its carbonic anhydrase inhibitory activity, a normal-anion-gap metabolic acidosis and polyuria may develop and persist for several days.[47]
Management of zonisamide overdose is entirely supportive. Key interventions include:
Given the exceptionally long elimination half-life of zonisamide, patients require a prolonged period of observation and supportive care until the drug is cleared and symptoms resolve.[54]
The effective and safe use of zonisamide in a clinical setting requires an individualized approach to therapy. Prescribing decisions must be guided by the drug's specific pharmacokinetic properties, its established efficacy data, and a comprehensive understanding of its potential risks. Dosing and titration strategies, in particular, must be adapted based on key patient-specific factors, most notably age and the use of concomitant medications that can alter its metabolism.
Zonisamide is available as hard capsules (Zonegran® and generics) in 25 mg, 50 mg, and 100 mg strengths, and as a 100 mg/5mL oral suspension (Zonisade™).[6] The capsules should be swallowed whole and not crushed or chewed.[8]
The speed of titration must be personalized. A slower titration schedule (e.g., bi-weekly increments) is appropriate for most patients, especially those not taking enzyme-inducing drugs or those with renal or hepatic impairment. Conversely, a faster titration (e.g., weekly increments) may be considered in patients taking potent CYP3A4-inducing AEDs, as they will clear zonisamide more rapidly.[2] This demonstrates that a clinician cannot use a rigid protocol; assessment of the patient's concomitant medication list is a critical prerequisite to designing an appropriate titration plan.
There is limited specific data on zonisamide use in the elderly. However, general geriatric prescribing principles apply. Dose selection should be cautious, typically starting at the lowest end of the dosing range, with a slower titration schedule. This is to account for the higher likelihood of age-related declines in renal, hepatic, and cardiac function, as well as the increased prevalence of polypharmacy.[2]
Zonisamide has secured a durable place in the therapeutic armamentarium for epilepsy as a second-generation antiepileptic drug. Its value lies in its unique 1,2-benzisoxazole structure and its multi-modal mechanism of action, which confers broad-spectrum efficacy, particularly as an adjunctive treatment for patients with refractory partial-onset seizures. The drug’s favorable pharmacokinetic profile, most notably its long elimination half-life, allows for convenient once- or twice-daily dosing schedules that can significantly enhance patient adherence.
However, the clinical utility of zonisamide is intrinsically linked to, and must be balanced against, a significant and mechanistically predictable safety profile. The primary risks are clustered around three core properties of the molecule: its sulfonamide chemical structure, which carries the potential for severe hypersensitivity reactions; its weak but clinically relevant inhibition of carbonic anhydrase, which predisposes patients to metabolic acidosis and nephrolithiasis; and its direct modulatory effects on the central nervous system, which can lead to cognitive side effects and an increased risk of suicidal ideation. Optimal use of zonisamide therefore demands careful patient selection, proactive monitoring for these specific risks, and comprehensive patient counseling.
Despite being on the market for several decades, zonisamide continues to be an active area of clinical and scientific investigation, with several unanswered questions and avenues for future development.
Published at: July 25, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.