Azetukalner is a novel, potent, and selective second-generation Kv7.2/7.3 potassium channel opener under late-stage clinical investigation by Xenon Pharmaceuticals.[1] It represents a significant advancement in a therapeutic class with a powerful, validated mechanism for modulating neuronal excitability. The drug is being developed in parallel for a range of high-unmet-need neurological and psychiatric conditions, including focal onset seizures (FOS), primary generalized tonic-clonic seizures (PGTCS), major depressive disorder (MDD), and bipolar depression (BPD).[1] This broad development program positions Azetukalner as a potential "pipeline-in-a-product" with transformative therapeutic potential.
In epilepsy, Azetukalner has demonstrated robust, dose-dependent efficacy in a Phase 2b trial for FOS.[4] More compellingly, long-term data from an ongoing 7-year open-label extension (OLE) study have shown sustained and deepening seizure reduction, with a median reduction in monthly seizure frequency exceeding 85% at 36 months.[5] The study has also yielded impressive rates of complete seizure freedom, with approximately one-third of patients in this highly treatment-resistant population achieving seizure freedom for a consecutive period of one year or longer.[6] These results suggest the potential for Azetukalner to shift the treatment paradigm for refractory epilepsy from seizure reduction to achievable seizure freedom.
In neuropsychiatry, a Phase 2 proof-of-concept study in MDD, while not meeting its primary endpoint, provided a strong signal of activity. The trial demonstrated a rapid onset of antidepressant effect, a statistically significant improvement in the core symptom of anhedonia, and a favorable outcome on a key secondary depression scale.[8] These findings, coupled with a safety profile that appears to circumvent common adverse effects of standard antidepressants such as weight gain and sexual dysfunction, have supported the drug's advancement into a comprehensive Phase 3 program for both MDD and BPD.[9]
Azetukalner's safety profile is a key differentiator. It was rationally designed to avoid the specific toxicity (blue skin discoloration) that led to the discontinuation of its predecessor, retigabine, and long-term clinical data have shown no evidence of this adverse event.[2] The overall profile is generally well-tolerated, with adverse events consistent with other anti-seizure medications.[4] With its novel mechanism, robust long-term efficacy in epilepsy, promising activity in depression, and favorable safety profile, Azetukalner is poised to become a significant, mechanistically distinct therapeutic option for a broad spectrum of central nervous system disorders.
Azetukalner is the officially adopted nonproprietary name for the investigational compound XEN1101, as approved by the United States Adopted Names (USAN) Council and the World Health Organization (WHO) International Nonproprietary Names (INN) expert committee.[10] Throughout its development, it has also been referred to by several other identifiers, including Encukalner, 1OP 2198, VRX 621698, and XPF-008.[1]
The compound was originally developed by Bausch Health Companies and is now under the stewardship of Xenon Pharmaceuticals Inc., a biopharmaceutical company focused on neuroscience.[1] Azetukalner is classified as a new molecular entity and represents a second-generation Kv7 potassium channel opener.[1] Its development was a direct result of a rational drug design strategy aimed at improving upon the first-generation Kv7 opener, retigabine. Retigabine, despite its efficacy, was ultimately withdrawn from the market due to serious side effects, most notably a blue-gray discoloration of the skin and eyes.[2] This toxicity was attributed to the in vivo formation of a chromophoric dimer originating from an aniline group in the retigabine structure. The medicinal chemistry program for Azetukalner specifically involved the removal of this reactive aniline moiety to eliminate the potential for such pigmentary changes, thereby de-risking the therapeutic class.[2] The success of this strategy has been borne out in clinical studies, where preliminary analyses from long-term open-label trials have revealed no reported pigmentary abnormalities, providing crucial clinical validation for this targeted molecular optimization.[11]
Azetukalner is a small molecule belonging to several chemical classes, including amides, anilides, and isoquinolines.[1] Its fundamental chemical identity and key properties are summarized in Table 1.
Table 1: Azetukalner Identification and Chemical Properties
| Identifier Type | Value | Source(s) |
|---|---|---|
| Official Name | Azetukalner | 12 |
| INN / USAN | azetukalner / AZETUKALNER | 10 |
| Company Code | XEN1101, XEN-1101 | 1 |
| Synonyms | Encukalner, 1OP-2198, VRX-621698, XPF-008 | 1 |
| Chemical Formula | $C_{23}H_{29}FN_{2}O$ | 12 |
| Molecular Weight | 368.49 g/mol | 2 |
| CAS Registry Number | 1009344-33-5 | 2 |
| PubChem CID | 24743936 | 13 |
| Systematic (IUPAC) Name | N-[4-(6-Fluoro-3,4-dihydro-2(1H)-isoquinolinyl)-2,6-dimethylphenyl]-3,3-dimethylbutanamide | 12 |
| SMILES | Cc1cc(cc(C)c1NC(=O)CC(C)(C)C)N2CCc3cc(ccc3C2)F | 2 |
| InChIKey | FJNPZKZPWVVSON-UHFFFAOYSA-N | 12 |
Voltage-gated potassium channels of the Kv7 family (encoded by the KCNQ genes, specifically KCNQ2 through KCNQ5) are fundamental regulators of neuronal activity in the central nervous system.[4] These channels generate a low-threshold, non-inactivating potassium current known as the M-current, which plays a crucial role in stabilizing the neuronal resting membrane potential and preventing excessive firing.[14] By facilitating the efflux of potassium ions ($K^+$), Kv7 channels help repolarize the neuron after an action potential, thereby exerting a powerful inhibitory control over neuronal firing.[18] This function is critical for preventing the kind of unwanted burst firing and spontaneous, hyperexcitable activity that underlies epileptic seizures.[18] The clinical relevance of this pathway is underscored by genetic evidence, as loss-of-function mutations in the genes encoding these channels are directly linked to human epilepsy syndromes.[4] Consequently, molecules that act as positive allosteric modulators, or "openers," of these channels represent a promising therapeutic strategy for disorders of neuronal hyperexcitability.[4]
Azetukalner functions as a novel, potent, and selective positive allosteric modulator of Kv7 potassium channels.[14] It specifically targets the heteromeric channels formed by Kv7.2 and Kv7.3 subunits, which are the predominant Kv7 subtypes expressed in the brain and are the primary contributors to the neuronal M-current.[2]
Potency: Azetukalner exhibits high potency in modulating channel activity. In vitro studies show a half-maximal effective concentration ($EC_{50}$) of 27 nM for opening Kv7.2/7.3 channels.[16] In functional patch-clamp assays, Azetukalner was found to be approximately 22 times more potent than the first-generation drug retigabine ($EC_{50}$ of 42 nM vs. 920 nM, respectively) in producing a leftward shift in the voltage of activation, a key indicator of enhanced channel opening at physiological membrane potentials.[16] This enhanced potency translates to in vivo activity, where Azetukalner was 16-fold more potent than retigabine in a rodent electroshock seizure model based on plasma concentrations ($EC_{50}$ of 220 nM vs. 3500 nM).[16]
Selectivity: The compound demonstrates notable selectivity for its target channels. It is approximately 4-fold more selective for the intended Kv7.2/7.3 heteromers over other Kv7 subtypes, such as Kv7.3/7.5 ($EC_{50}$ = 94 nM) and Kv7.4 ($EC_{50}$ = 113 nM).[16] Furthermore, Azetukalner exhibits a selectivity of over 100-fold for Kv7 channels compared to a broad panel of other ion channels and receptors, which likely contributes to its generally favorable tolerability profile by minimizing off-target effects.[16]
The drug's mechanism as a neuronal circuit stabilizer provides a compelling pharmacological basis for its investigation across different CNS disorders. In epilepsy, a condition defined by hyperexcitability, its action of enhancing an inhibitory potassium current directly counteracts the underlying pathophysiology.[4] In major depressive disorder, where dysregulation of neural circuits is also implicated, the ability to dampen excessive neuronal activity in key mood-regulating pathways presents a novel antidepressant mechanism. The observed efficacy against anhedonia, in particular, may relate to the modulation of reward circuits, which are also governed by principles of neuronal excitability.[8] This unified mechanism suggests Azetukalner is not merely an anti-seizure drug with mood effects, but rather a fundamental neuronal stabilizer with broad therapeutic potential.
Azetukalner is an orally bioavailable small molecule formulated for convenient once-daily (QD) administration.[2] Clinical trial protocols consistently specify that the drug should be taken with food, typically with an evening meal.[18] A significant practical advantage of its pharmacokinetic profile is the lack of a requirement for dose titration at the beginning of treatment or tapering upon cessation.[4] This simplifies the dosing regimen for both physicians and patients, potentially improving treatment adherence.
The primary metabolic pathway for Azetukalner involves the Cytochrome P450 3A4 (CYP3A4) enzyme system.[11] This reliance on a single, major metabolic enzyme has important clinical implications for potential drug-drug interactions, which are discussed in a later section.
Azetukalner has undergone extensive clinical investigation in epilepsy, with a robust program that has established a strong foundation of efficacy and safety data. The overall clinical development program is summarized in Table 2.
Table 2: Overview of Key Azetukalner Clinical Trials
| Trial Name (NCT ID) | Indication | Phase | Status | Primary Endpoint | Source(s) |
|---|---|---|---|---|---|
| X-TOLE (NCT03796962) | Focal Onset Seizures (FOS) | 2b | Completed | Median % change in monthly FOS frequency | 4 |
| X-TOLE OLE | FOS (Long-term) | OLE | Ongoing | Long-term safety and efficacy | 4 |
| X-TOLE2 (NCT05614063) | FOS | 3 | Ongoing | Median % change in monthly FOS frequency | 19 |
| X-TOLE3 | FOS | 3 | Ongoing | Median % change in monthly FOS frequency | 19 |
| X-ACKT | PGTCS | 3 | Ongoing | Median % change in monthly PGTCS frequency | 10 |
| X-NOVA (NCT05376150) | Major Depressive Disorder (MDD) | 2 | Completed | Change in MADRS score at Week 6 | 8 |
| X-NOVA2 | MDD | 3 | Initiated | Change in HAM-D17 score at Week 6 | 23 |
| X-NOVA3 (NCT07076407) | MDD | 3 | Recruiting | Change in HAM-D17 score at Week 6 | 1 |
| X-CEED (NCT07172516) | Bipolar Depression (BPD) | 3 | Recruiting | Change in MADRS score at Week 6 | 24 |
The X-TOLE study was a pivotal Phase 2b randomized, double-blind, placebo-controlled trial that provided the initial proof-of-concept for Azetukalner in epilepsy. The study evaluated three once-daily doses (10 mg, 20 mg, and 25 mg) administered with food against placebo as adjunctive therapy in adults with FOS.[4] The trial successfully met its primary endpoint, demonstrating a statistically significant and dose-dependent reduction in monthly FOS frequency from baseline over an 8-week treatment period. The median percentage reductions were 33.2% ($p=0.04$), 46.4% ($p<0.001$), and 52.8% ($p<0.001$) for the 10 mg, 20 mg, and 25 mg groups, respectively, compared to an 18.2% reduction in the placebo group.[4] A particularly noteworthy finding was the drug's rapid onset of action, with statistically significant reductions in seizure frequency observed within the first week of treatment across all dose groups.[4]
Following the double-blind phase, 275 eligible participants (96.5%) enrolled in an ongoing 7-year OLE study, in which all patients receive Azetukalner 20 mg once daily with food.[4] This study has generated a substantial body of long-term data, with over 700 patient-years of exposure and some individuals remaining on treatment for more than five years.[6] The OLE has provided compelling evidence of the drug's durable efficacy and long-term safety.
Sustained Efficacy: The robust anti-seizure effect observed in the X-TOLE trial was not only sustained but appeared to deepen over time in the OLE. Interim data analyses have consistently shown profound reductions in seizure frequency from the original study baseline. The median percentage change (MPC) in monthly FOS frequency was -83.2% at 24 months, -86.9% at 30 months, and approximately -85% at 36 months of OLE treatment.[5]
Seizure Freedom: The most clinically impactful finding from the OLE has been the high rate of complete seizure freedom achieved in this difficult-to-treat patient population. The participants entering the study had a history of refractory epilepsy, having tried and discontinued a median of six prior anti-seizure medications (ASMs), with over half taking three concomitant ASMs at baseline.[4] Despite this high level of treatment resistance, a significant portion of patients achieved prolonged periods of seizure freedom. For those who had been treated for at least 24 months, 23.6% achieved seizure freedom for a consecutive period of 12 months or more.[4] This rate increased further with longer treatment duration; among patients treated for at least 36 months, approximately one-third (32.7%) achieved seizure freedom for at least 12 consecutive months.[6] This level of efficacy in a heavily pre-treated population suggests a powerful and durable mechanistic effect that could potentially shift the treatment goal for refractory FOS from seizure reduction to attainable seizure freedom, a metric that directly translates to improved quality of life.[6]
Durability and Retention: The long-term tolerability and efficacy are further supported by strong patient retention rates. In the OLE, retention was 66% at 12 months, 60% at 24 months, and 52% at 36 months, which is impressive for a long-term trial in this patient population.[4]
Based on the strong Phase 2b and OLE results, Xenon has initiated a pivotal Phase 3 program for FOS, consisting of two identical trials: X-TOLE2 (NCT05614063) and X-TOLE3.[18] These multicenter, randomized, double-blind, placebo-controlled trials are designed to support regulatory submissions for approval. Each trial aims to enroll approximately 360 adults with FOS who are on a stable regimen of 1 to 3 background ASMs.[19] Participants are randomized in a 1:1:1 ratio to receive either Azetukalner 15 mg, Azetukalner 25 mg, or placebo, administered once daily with food for a 12-week double-blind treatment period.[19] The primary efficacy endpoint for both trials is the median percent change in monthly focal seizure frequency from baseline compared to placebo.[19]
To broaden the potential utility of Azetukalner in epilepsy, the company is also conducting the Phase 3 X-ACKT trial, which is intended to support a potential label expansion for the treatment of PGTCS.[3] This multicenter, randomized, double-blind, placebo-controlled study is planned to enroll approximately 160 patients aged 12 years and older with PGTCS.[18] Participants will be randomized 1:1 to receive either Azetukalner 25 mg or placebo once daily with food as an adjunctive treatment. The primary endpoint is the median percent change in monthly PGTCS frequency from baseline.[18]
The X-NOVA trial was a Phase 2, multicenter, randomized, double-blind, placebo-controlled study designed to evaluate the efficacy and safety of Azetukalner as a monotherapy for MDD.[8] The trial enrolled 168 adults with moderate-to-severe MDD, who were randomized to receive Azetukalner 10 mg, Azetukalner 20 mg, or placebo once daily for six weeks.[9]
Primary and Secondary Endpoints: The study did not meet its pre-specified primary endpoint, which was a statistically significant change from baseline in the Montgomery-Åsberg Depression Rating Scale (MADRS) total score at week 6.[8] However, the 20 mg dose group demonstrated a clear dose response and a clinically meaningful, though not statistically significant, difference of -3.04 points versus the placebo group ($p=0.135$).[9]
Despite the primary endpoint result, the trial yielded several statistically significant findings that provided a strong signal of drug activity and supported further development. These included:
The decision to advance to Phase 3 was based on this totality of evidence. The combination of a clinically meaningful effect on the primary endpoint, coupled with statistically significant results on a key secondary depression scale, rapid onset of action, and a novel effect on anhedonia, provided a compelling rationale. This interpretation was validated by the U.S. Food and Drug Administration (FDA), which agreed to the Phase 3 program following an "end-of-Phase 2" meeting.[10] This suggests that Azetukalner's potential in MDD may lie in a differentiated clinical profile rather than simply replicating the efficacy of existing treatments.
Based on the X-NOVA results and alignment with the FDA, Xenon has initiated a comprehensive Phase 3 program for MDD.[10] The program is planned to include three multicenter, randomized, double-blind, placebo-controlled trials. The first two trials, X-NOVA2 and X-NOVA3 (NCT07076407), have been initiated.[1] These trials will evaluate Azetukalner 20 mg once daily with food as a monotherapy over a 6-week period.[23] Reflecting the findings from the Phase 2 study, the primary efficacy endpoint for the Phase 3 program has been designated as the change from baseline in the HAM-D17 score at week 6.[23]
The neuropsychiatric development of Azetukalner has been expanded to include bipolar depression, a condition with significant unmet medical need. The X-CEED trial is the first of two planned Phase 3 studies for this indication.[24] It is a multicenter, randomized, double-blind, placebo-controlled study designed to enroll approximately 400 adult participants diagnosed with Bipolar I or Bipolar II disorder who are currently experiencing a major depressive episode.[24] Participants will be randomized to receive either Azetukalner 20 mg once daily with food or placebo for a 6-week treatment period.[24] The primary efficacy endpoint is the change from baseline in the MADRS total score at week 6.[24]
Key inclusion criteria for the X-CEED trial include an age range of 18 to 74 years, a formal DSM-5-TR diagnosis of Bipolar I or II disorder, and a current major depressive episode with a duration of 4 to 52 weeks.[24] Key exclusion criteria are designed to ensure a homogenous patient population and patient safety, and include treatment resistance in the current depressive episode, active suicidality, a Young Mania Rating Scale (YMRS) score >12 (to exclude patients with manic or hypomanic symptoms), and recent use of other psychotropic medications.[24]
Across its extensive clinical development program, Azetukalner has been consistently described as generally well tolerated.[4] The profile of treatment-emergent adverse events (TEAEs) is consistent with that of other commonly prescribed anti-seizure medications, and importantly, no new safety signals have been identified during long-term exposure in the OLE studies.[4] A consolidated summary of the most common TEAEs from the two largest reported studies is provided in Table 3, allowing for a comparison of the safety profile in both epilepsy and MDD populations.
Table 3: Summary of Common Treatment-Emergent Adverse Events (TEAEs) Across Key Studies
| Adverse Event | X-TOLE OLE (Epilepsy, N=275) | X-NOVA (MDD, 20 mg, N=56) | X-NOVA (MDD, Placebo, N=56) |
|---|---|---|---|
| Dizziness | 22.5% | 17.9% | 7.3% |
| Somnolence (Sleepiness) | 13.8% | 10.7% | 1.8% |
| Headache | 16.0% | 8.9% | 12.7% |
| Disturbance in Attention | 5.5% | 8.9% | 0% |
| Fall | 13.1% | Not Reported | Not Reported |
| Memory Impairment | 11.6% | Not Reported | Not Reported |
| Coronavirus Infection | 16.4% | Not Reported | Not Reported |
| Weight Increase | 10.5% | Not Reported | Not Reported |
Note: Data sourced from.[4] "Not Reported" indicates the event was not listed among the most common TEAEs for that study arm.
In the long-term X-TOLE OLE study for epilepsy, the majority of TEAEs were mild or moderate in severity.[5] The most frequently reported TEAEs (occurring in ≥10% of participants) were dizziness (22.5%), coronavirus infection (16.4%), headache (16.0%), somnolence (13.8%), fall (13.1%), memory impairment (11.6%), and weight increase (10.5%).[5] Serious adverse events (SAEs) were reported in 14.2% of participants, with 2.2% considered treatment-related. The rate of discontinuation due to an adverse event was 12.4%.[5]
In the 6-week X-NOVA study for MDD, Azetukalner was also well tolerated. In the 20 mg dose group, the most common TEAEs were primarily related to the central nervous system and included dizziness (17.9%), somnolence (10.7%), headache (8.9%), and disturbance in attention (8.9%).[9] Importantly, rates of discontinuation due to TEAEs were low and comparable to placebo (5.4% for the 20 mg group vs. 3.6% for placebo).[9]
Azetukalner's safety profile appears strategically advantageous for two key reasons. First, as a second-generation Kv7 opener, it was specifically engineered to avoid the mechanism-related toxicity of retigabine. Long-term clinical data have successfully validated this approach, with no reports of the pigmentary abnormalities that plagued its predecessor.[2] This effectively overcomes a major historical barrier for the therapeutic class.
Second, in the context of depression, the drug appears to circumvent some of the most common and burdensome side effects of standard-of-care antidepressants. Data from the X-NOVA study showed that Azetukalner was not associated with notable weight gain or sexual dysfunction, two adverse effects that are major drivers of non-adherence to SSRI and SNRI therapies.[9] This dual safety advantage—solving an inherent class toxicity while also offering a potentially better-tolerated alternative to existing standards of care—could be a significant driver of clinical adoption if the drug receives regulatory approval.
The primary route of metabolism for Azetukalner is via the cytochrome P450 3A4 (CYP3A4) enzyme.[11] This is one of the most important and common metabolic pathways for xenobiotics, meaning Azetukalner is susceptible to a wide range of potential drug-drug interactions (DDIs).[32] The co-administration of drugs that strongly modulate CYP3A4 activity can significantly alter Azetukalner's plasma exposure.[11]
Given this well-defined metabolic pathway, it is highly probable that any future product labeling for Azetukalner will include specific warnings and dose adjustment recommendations for its use with strong CYP3A4 modulators. This is a critical consideration for ensuring the safe and effective clinical use of the drug, particularly in patient populations prone to polypharmacy.
As Azetukalner is still an investigational agent, no formal contraindications have been established by regulatory authorities.[26] However, the inclusion and exclusion criteria from its clinical trials provide valuable insight into populations where its use has not been studied or where caution may be warranted. For instance, the Phase 3 trial in bipolar depression (X-CEED) excludes individuals with a recent history of active suicidal plans or behavior, current substance use disorders, or a Young Mania Rating Scale score indicative of mania or hypomania.[24] Similarly, the Phase 3 epilepsy trials (X-TOLE2/3) exclude patients whose seizures are secondary to other acute or progressive conditions (such as alcohol withdrawal, infection, or neoplasia) and those with a recent history of status epilepticus.[29]
Azetukalner is a late-stage investigational drug that has not been approved for marketing by the FDA or any other global regulatory agency.[18] It is actively being evaluated in a comprehensive Phase 3 clinical development program across four major indications: focal onset seizures, primary generalized tonic-clonic seizures, major depressive disorder, and bipolar depression.[1] Xenon anticipates that enrollment in the first Phase 3 FOS trial (X-TOLE2) will be completed in late 2024 to early 2025.[10]
Xenon Pharmaceuticals has engaged with the FDA at key stages of development. Most notably, the company held a successful "end-of-Phase 2" meeting with the FDA for the MDD program, which resulted in alignment on the key design features of the pivotal Phase 3 program, allowing it to proceed.[10] In a standard step for a drug advancing toward regulatory submission, the compound has been assigned the official nonproprietary name "azetukalner" by both USAN and WHO.[10] There is currently no available information regarding the drug's regulatory status or submissions to other health authorities, such as Australia's Therapeutic Goods Administration (TGA).[1]
Azetukalner is currently positioned as the most advanced and clinically validated Kv7 potassium channel modulator in late-stage development.[3] If approved, it has the potential to be the only-in-class selective Kv7.2/7.3 opener available on the market, offering a novel and structurally distinct mechanism of action compared to all currently marketed ASMs and antidepressants.[11]
The drug's future outlook is exceptionally strong, underpinned by a broad, multi-indication development strategy that diversifies risk and maximizes commercial potential. This "pipeline-in-a-product" approach is supported by two pillars of evidence:
Ultimately, Azetukalner represents more than just a single drug candidate; it is the clinical validation of a powerful therapeutic hypothesis: that stabilizing neuronal excitability through the targeted modulation of Kv7.2/7.3 channels has broad utility across a spectrum of CNS disorders. Its trajectory toward becoming a cornerstone therapy in both neurology and psychiatry will depend on the successful execution of its ongoing pivotal trials and subsequent regulatory reviews.
Azetukalner (XEN1101) has emerged as a highly promising, late-stage therapeutic candidate with a unique and compelling profile. As a second-generation Kv7.2/7.3 potassium channel opener, it leverages a potent mechanism for neuronal stabilization while successfully overcoming the key toxicity that limited its predecessor. The extensive clinical data gathered to date, particularly the remarkable long-term efficacy and seizure freedom rates observed in treatment-resistant focal epilepsy, strongly support its potential to become a paradigm-shifting anti-seizure medication.
Furthermore, the parallel expansion into major psychiatric disorders is supported by promising Phase 2 data in MDD, which highlighted a differentiated profile characterized by rapid onset, a significant impact on anhedonia, and a favorable safety profile that avoids common side effects of standard antidepressants. The ongoing Phase 3 programs in FOS, PGTCS, MDD, and BPD underscore the broad potential of its underlying mechanism. With a convenient once-daily dosing regimen without the need for titration and a generally well-tolerated safety profile, Azetukalner is well-positioned to address significant unmet medical needs across both neurology and psychiatry, potentially establishing a new and valuable class of CNS therapeutics.
Published at: October 17, 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.