C20H25NO2S2
115103-54-3
Partial-Onset Seizures
Tiagabine is an anti-epileptic drug (AED) distinguished by its unique and highly specific mechanism of action: the selective inhibition of the gamma-aminobutyric acid (GABA) transporter 1 (GAT-1).[1] This mechanism enhances GABAergic neurotransmission, positioning Tiagabine as a targeted therapy for controlling neuronal hyperexcitability. Its primary, United States Food and Drug Administration (FDA)-approved clinical indication is as an adjunctive therapy for the treatment of partial-onset seizures in adults and adolescents aged 12 years and older.[3]
The clinical application of Tiagabine is fundamentally governed by a critical pharmacokinetic dichotomy: its metabolism is significantly influenced by the presence of hepatic enzyme-inducing concomitant medications. Patients co-administered enzyme-inducing AEDs (e.g., carbamazepine, phenytoin) exhibit markedly accelerated clearance of Tiagabine compared to "non-induced" patients. This metabolic variability necessitates distinct dosing and titration strategies for these two populations to ensure both efficacy and safety. While the drug has been explored for off-label uses, such as in anxiety disorders and neuropathic pain, its use in non-epileptic populations has been associated with significant safety concerns, including the paradoxical development of new-onset seizures, which has led to prominent regulatory warnings.[5] This report provides a comprehensive monograph on Tiagabine, detailing its chemical properties, pharmacological profile, clinical applications, safety considerations, and regulatory history.
A precise and unambiguous identification of Tiagabine is essential for research, clinical, and regulatory purposes. The drug is most commonly administered as a hydrochloride salt, a distinction that is critical for understanding its physicochemical properties and formulation. The following table consolidates its nomenclature and cross-references from major chemical and pharmacological databases.
| Identifier Type | Value | Source(s) |
|---|---|---|
| Generic Name | Tiagabine | 7 |
| International Names | Tiagabina (Spanish), Tiagabinum (Latin) | 8 |
| Brand Name | Gabitril | 3 |
| DrugBank Accession No. | DB00906 | 7 |
| CAS Registry Number | 115103-54-3 (free base) | 10 |
| 145821-59-6 (hydrochloride salt) | 12 | |
| Drug Type/Modality | Small Molecule | 7 |
| Systematic (IUPAC) Name | (3R)-1-[4,4-bis(3-methylthiophen-2-yl)but-3-enyl]piperidine-3-carboxylic acid | 10 |
| Chemical Name (HCl Salt) | (-)-(R)-1-nipecotic acid hydrochloride | 15 |
| Synonyms/Dev. Codes | TGB, NO-05-0328, NNC-05-0328, Abbott-70569, ABT-569, NO-328 | 14 |
| FDA UNII | Z80I64HMNP | 10 |
| ChEBI ID | CHEBI:9586 | 10 |
| ChEMBL ID | CHEMBL1027 | 10 |
| KEGG ID | D08588, C07503 | 10 |
| PubChem CID | 60648 | 14 |
| ATC Code | N03AG06 | 3 |
Tiagabine is a synthetic organic compound derived from (R)-nipecotic acid, a piperidinemonocarboxylic acid.[10] Its structure incorporates a lipophilic 1,1-bis(3-methyl-2-thienyl)but-1-en-4-yl group attached to the nitrogen of the nipecotic acid ring, which is crucial for its ability to cross the blood-brain barrier.[3]
Tiagabine is formulated for oral administration as immediate-release tablets under the brand name Gabitril.[15]
The formulation and administration guidelines for Tiagabine are closely linked to its physicochemical and pharmacokinetic properties. The drug exhibits high overall absorption (>95%), but its rate of absorption is significantly affected by food.[7] Administration with a high-fat meal can delay the time to reach peak plasma concentration (Tmax) from approximately 45 minutes to 2.5 hours.[3] This slowing of the absorption rate, without altering the total amount of drug absorbed, serves a critical clinical purpose. The most common adverse effects of Tiagabine, such as dizziness and somnolence, are dose-related and linked to peak plasma concentrations.[1] By recommending that the drug be taken with food, the resulting blunted peak concentration helps to improve patient tolerability, particularly during the initial dose-titration period when the central nervous system is adapting to the medication's effects. This administration guideline is therefore a key strategy for mitigating predictable, concentration-dependent side effects.
The anticonvulsant effect of Tiagabine is attributed to its potent and selective action as a GABA reuptake inhibitor.[7] While some clinical literature refers to the "precise mechanism" of its antiseizure effect as unknown, this reflects the complexity of translating a molecular action into a network-level clinical outcome in epilepsy; the primary pharmacological mechanism is well-characterized.[7]
Tiagabine's primary molecular target is the Sodium- and chloride-dependent GABA transporter 1 (GAT-1).[2] GAT-1 is a key presynaptic and glial membrane protein responsible for the rapid removal of GABA from the synaptic cleft following its release, thereby terminating its inhibitory signal.[17] By binding to recognition sites on the GAT-1 carrier, Tiagabine potently blocks this reuptake process.[10] This inhibition leads to an increased concentration of GABA in the synapse and prolongs its duration of action on postsynaptic GABAA and GABAB receptors.[3] This enhancement of the brain's primary inhibitory neurotransmitter system is believed to counteract the neuronal hyperexcitability that underlies seizure generation and propagation.[15] In vitro and in vivo studies have confirmed this mechanism, demonstrating that Tiagabine prolongs GABA-mediated inhibitory post-synaptic potentials (IPSPs) and increases extracellular GABA levels in key brain regions such as the hippocampus, globus pallidus, and substantia nigra.[3]
A defining feature of Tiagabine is its high selectivity for the GAT-1 transporter, which contributes to a relatively clean neurological side-effect profile compared to less selective agents.[17] It has minimal activity at other neurotransmitter transporters or receptors at clinically relevant concentrations.
The interaction of Tiagabine with GAT-1 is structurally specific. The nipecotic acid portion of the molecule occupies the primary GABA binding site, while the lipophilic bis-thienyl side chains interact with an allosteric site, effectively locking the transporter in an outward-facing, non-transporting conformation.[3] This lipophilicity is a key design feature that allows the molecule to effectively penetrate the blood-brain barrier, unlike its parent compound, nipecotic acid.[3] Furthermore, preclinical evidence suggests that Tiagabine may indirectly potentiate the effects of other GABAergic drugs, as it has been shown to increase the affinity of benzodiazepines for the GABAA receptor complex.[3]
The pharmacokinetics of Tiagabine are well-characterized and linear within the therapeutic dose range. However, its clinical use is dominated by its susceptibility to metabolic induction by concomitant medications, creating two distinct patient phenotypes: "induced" and "non-induced."
Tiagabine is rapidly and almost completely absorbed following oral administration, with an absolute bioavailability exceeding 95%.[3] Under fasting conditions, peak plasma concentrations (Tmax) are reached within approximately 45 minutes.[3] As previously noted, co-administration with food slows the rate of absorption but does not impact the overall extent.[16] Steady-state plasma concentrations are typically achieved within two days of initiating a stable dosing regimen.[15]
Tiagabine is extensively bound to plasma proteins (approximately 96%), primarily to serum albumin and α1-acid glycoprotein.[7] This high degree of protein binding suggests a potential for displacement interactions with other highly protein-bound drugs, although the clinical significance of this is managed through careful dosing.
Tiagabine is extensively metabolized by the liver, with less than 2% of the dose excreted unchanged.[15] The primary metabolic pathway involves oxidation of the thiophene rings, a reaction mediated predominantly by the cytochrome P450 3A4 (CYP3A4) isoenzyme.[3] This process yields pharmacologically inactive metabolites, such as 5-oxo-tiagabine.[3] A secondary metabolic route is glucuronidation.[3] While minor contributions from other CYP isoforms (CYP1A2, CYP2D6, CYP2C19) cannot be entirely ruled out, CYP3A4 is the principal enzyme responsible for Tiagabine clearance.[15]
The elimination of Tiagabine is highly dependent on the patient's enzyme-induction status.
This profound difference in metabolic rate is the single most important pharmacokinetic consideration in the clinical use of Tiagabine. It means that for a given dose, plasma concentrations in non-induced patients can be more than double those seen in induced patients.[15] This reality underpins the need for separate dosing guidelines and explains the heightened risk of adverse events when the drug is used in non-induced populations, particularly for off-label indications. Following metabolism, the byproducts are primarily excreted in the feces (approximately 63%) and urine (approximately 25%).[7]
The following table summarizes and contrasts the key pharmacokinetic parameters of Tiagabine in these two distinct patient populations.
| PK Parameter | Value in Non-Induced Patients | Value in Induced Patients | Key Notes | Source(s) |
|---|---|---|---|---|
| Bioavailability | >95% | >95% | High and complete absorption, unaffected by induction status. | 3 |
| Tmax (Fasting) | ~45 minutes | ~45 minutes | Rate of absorption is not affected by induction status. | 3 |
| Protein Binding | ~96% | ~96% | Primarily to albumin and α1-acid glycoprotein. | 7 |
| Primary Metabolism | Hepatic (CYP3A4) | Hepatic (CYP3A4) | Metabolism is dramatically accelerated by CYP3A4 inducers. | 7 |
| Elimination Half-Life | 7–9 hours | 2–5 hours | Reduced by 50-65% due to enzyme induction. This is the critical difference. | 3 |
| Excretion | 25% renal, 63% fecal (as metabolites) | 25% renal, 63% fecal (as metabolites) | Excretion route is unchanged, but rate of metabolite formation is faster. | 3 |
The sole FDA-approved indication for Tiagabine is for use as an adjunctive therapy in the treatment of partial-onset seizures in adults and adolescents aged 12 years and older.[3] This includes seizure types classified as focal aware (formerly simple partial), focal impaired awareness (formerly complex partial), and focal to bilateral tonic-clonic (formerly secondarily generalized) seizures.[1] It is specifically intended as an "add-on" medication for patients whose seizures are not satisfactorily controlled with other AEDs.[9]
The efficacy of Tiagabine for this indication was established in three pivotal multicenter, double-blind, placebo-controlled clinical trials. These studies enrolled a total of 769 patients with refractory partial seizures who, critically, were already being treated with at least one hepatic enzyme-inducing AED.[15] The results demonstrated that Tiagabine, at maintenance doses between 30 and 56 mg/day, was significantly more effective than placebo in reducing seizure frequency across all subtypes of partial seizures.[1] Furthermore, long-term safety studies have shown that the efficacy of Tiagabine is sustained over time, with no evidence of pharmacological tolerance developing.[1] Some open-label extension studies have also suggested that a subset of patients may be successfully converted from polytherapy to Tiagabine monotherapy, although it is not formally approved for this use.[1]
The mechanism of Tiagabine—enhancing central GABAergic inhibition—has made it an attractive candidate for conditions beyond epilepsy that are hypothesized to involve GABAergic dysregulation. However, its off-label use is accompanied by a significant and specific safety risk.
The history of Tiagabine's off-label use serves as a critical case study in clinical pharmacology. The drug's initial safety and dosing profile was established in a population of epilepsy patients who were concurrently taking enzyme-inducing AEDs. When clinicians began prescribing Tiagabine for psychiatric or pain conditions in patients who were not taking these inducers, they were inadvertently treating a "non-induced" population. As established, these individuals metabolize Tiagabine much more slowly, leading to plasma concentrations more than twice as high as those observed in the original trial population for the same dose.[15] This unexpectedly high drug exposure in individuals without an underlying epileptic focus led to a paradoxical pro-convulsant effect, resulting in reports of new-onset seizures and status epilepticus.[5] This prompted the FDA to issue a prominent, bolded warning in 2005, cautioning against the use of Tiagabine in patients without epilepsy due to this risk.[5] This warning fundamentally limits the off-label utility of the drug and underscores the necessity of understanding its pharmacokinetic profile before prescribing.
The safe and effective use of Tiagabine is highly dependent on strict adherence to established dosing and administration protocols, which are designed to mitigate its dose-related adverse effects and account for its variable metabolism.
The following guidelines apply to all patients receiving Tiagabine:
Dosage and titration schedules for Tiagabine must be tailored based on the patient's concomitant medications, specifically whether they are taking hepatic enzyme-inducing AEDs.
This population includes patients taking medications such as carbamazepine, phenytoin, phenobarbital, and primidone. They require higher maintenance doses due to accelerated drug clearance.
The following table provides a typical titration regimen for induced patients.
| Week of Therapy | Action | Total Daily Dose | Dosing Frequency |
|---|---|---|---|
| Week 1 | Initiate at 4 mg once daily | 4 mg/day | Once daily |
| Week 2 | Increase total daily dose by 4 mg | 8 mg/day | Two divided doses |
| Week 3 | Increase total daily dose by 4 mg | 12 mg/day | Three divided doses |
| Week 4 | Increase total daily dose by 4 mg | 16 mg/day | Two to four divided doses |
| Week 5 | Increase total daily dose by 4 to 8 mg | 20 to 24 mg/day | Two to four divided doses |
| Week 6 | Increase total daily dose by 4 to 8 mg | 24 to 32 mg/day | Two to four divided doses |
| Maintenance | Titrate to clinical response | 32 to 56 mg/day | Two to four divided doses |
This population experiences more than double the plasma concentration for a given dose compared to induced patients. Consequently, treatment requires significantly lower doses and a much slower rate of titration.[15] While a specific, universally accepted titration schedule for this group is not as rigorously defined as for induced patients, the guiding principle is extreme caution, with smaller initial doses and longer intervals between dose increases.
The safety profile of Tiagabine is characterized primarily by dose-related central nervous system effects and a significant risk of paradoxical seizures when used inappropriately, particularly in non-epileptic individuals.
The following table summarizes the adverse effects associated with Tiagabine, categorized by system organ class.
| System Organ Class | Adverse Effect | Typical Severity/Frequency | Source(s) |
|---|---|---|---|
| Nervous System | Dizziness, Somnolence/Drowsiness | Very Common; dose-related, often during titration | 1 |
| Asthenia (Weakness/Lack of Energy) | Common; can be incapacitating in some cases | 1 | |
| Tremor | Common | 21 | |
| Difficulty with Concentration/Attention | Common | 21 | |
| Speech/Language Problems, Confusion | Less Common; may be associated with EEG changes | 29 | |
| New-onset Seizures / Status Epilepticus | Rare but Serious; primarily in off-label use in non-epileptics | 5 | |
| Non-convulsive Status Epilepticus | Rare but Serious; can occur in patients with epilepsy | 4 | |
| Psychiatric | Nervousness/Irritability | Common | 1 |
| Depression | Less Common; reported in ~4% of epilepsy patients | 17 | |
| Suicidal Ideation and Behavior | Rare but Serious; class effect for all AEDs | 9 | |
| Gastrointestinal | Nausea | Common | 1 |
| Abdominal Pain, Diarrhea | Less Common | 15 | |
| Skin and Subcutaneous Tissue | Rash | Less Common | 30 |
| Stevens-Johnson Syndrome (SJS), Toxic Epidermal Necrolysis (TEN) | Very Rare but Life-Threatening | 32 |
Notably, unlike some other AEDs, Tiagabine therapy has not been associated with clinically significant serum aminotransferase elevations or hepatotoxicity.[21]
Tiagabine is contraindicated in the following situations:
The potential for drug-drug interactions with Tiagabine is primarily driven by its metabolism via CYP3A4 and its intrinsic CNS depressant effects. The following table outlines the most clinically significant interactions.
| Interacting Drug/Class | Mechanism of Interaction | Effect on Tiagabine | Effect on Other Drug | Clinical Recommendation | Source(s) |
|---|---|---|---|---|---|
| Enzyme-Inducing AEDs (Carbamazepine, Phenytoin, Phenobarbital, Primidone) | CYP3A4 Induction | Decreased plasma concentration and half-life (~50-65%) | None (Tiagabine does not alter their levels) | Higher doses of Tiagabine are required. Follow "Induced Patient" dosing guidelines. | 23 |
| Other CYP3A4 Inducers (e.g., Rifampin, St. John's Wort, Bosentan) | CYP3A4 Induction | Decreased plasma concentration | Variable | Monitor for reduced Tiagabine efficacy; dose increase may be needed. | 20 |
| CYP3A4 Inhibitors (e.g., Ketoconazole, Clarithromycin, Ritonavir, Nefazodone) | CYP3A4 Inhibition | Increased plasma concentration | Variable | Monitor for Tiagabine toxicity; dose reduction may be necessary. | 7 |
| CNS Depressants (Alcohol, Benzodiazepines, Opioids, Antihistamines, Antipsychotics) | Additive Pharmacodynamic Effect | None | Increased sedation, dizziness, cognitive and motor impairment | Counsel patient on the risk of additive CNS depression. Avoid co-use with alcohol. | 7 |
| Valproate | Protein Binding Displacement | May increase free (unbound) Tiagabine levels | None | Monitor for increased Tiagabine side effects. | 32 |
| Drugs that Lower Seizure Threshold (e.g., Amphetamine, Chloroquine, Mefloquine) | Pharmacodynamic Antagonism | Decreased therapeutic efficacy | Variable | Co-administration may reduce anticonvulsant effect. Use with caution. | 7 |
An important clinical consideration is the largely unidirectional nature of its pharmacokinetic interactions with other major AEDs. While enzyme inducers like carbamazepine and phenytoin profoundly reduce Tiagabine levels, studies have shown that the addition of Tiagabine to a stable regimen of these drugs does not significantly alter their steady-state concentrations.[36] This simplifies therapeutic drug management, as clinicians can focus on adjusting the Tiagabine dose without needing to preemptively modify the dose of the concomitant inducer.
Tiagabine represents a mechanistically targeted therapeutic agent in the field of epileptology. Its high selectivity as an inhibitor of the GAT-1 GABA transporter offers a precise method for enhancing synaptic inhibition, a fundamental strategy for seizure control. Its efficacy as an add-on therapy for refractory partial-onset seizures is supported by robust clinical trial data.
However, the clinical utility of Tiagabine is sharply defined and constrained by its pharmacokinetic profile. The drug's heavy reliance on the CYP3A4 metabolic pathway makes its clearance exceptionally sensitive to the presence of enzyme-inducing or -inhibiting drugs. This creates a stark dichotomy between "induced" and "non-induced" patient populations, demanding fundamentally different clinical management approaches. The history of its off-label use, which led to paradoxical seizures in non-induced individuals, serves as a powerful illustration of how a failure to appreciate these pharmacokinetic principles can transform a therapeutic agent into a source of significant risk. The resulting regulatory warnings have appropriately narrowed its clinical application, reinforcing its role as a specialized tool rather than a broad-spectrum agent.
The paramount consideration for any clinician prescribing Tiagabine is the patient's co-medication profile. A thorough assessment of a patient's enzyme induction status is not merely advisable but essential for safe and effective dosing. The principles of slow, careful dose titration and administration with food must be strictly followed to enhance tolerability.
Tiagabine's journey from development to post-marketing surveillance offers valuable lessons for clinical pharmacology. It underscores that a drug's safety profile is not an intrinsic property alone but an emergent one, arising from the complex interplay between its mechanism, its pharmacokinetics, and the specific patient population in which it is used. While its broader application has been limited, Tiagabine remains a viable option for appropriately selected patients with refractory partial epilepsy. Future research could explore novel drug delivery systems that might provide more consistent plasma levels, potentially mitigating the risks associated with its variable metabolism and widening its therapeutic index.
Published at: October 19, 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.