RAP-219 is a clinical-stage, investigational small molecule being developed by Rapport Therapeutics as a potential first-in-class precision medicine for neurological and psychiatric disorders. Its primary mechanism of action is as a highly selective negative allosteric modulator (NAM) of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors that are specifically associated with the Transmembrane AMPA Receptor Regulatory Protein γ8 (TARPγ8). This mechanism confers neuroanatomical specificity, as TARPγ8 is enriched in brain regions critical to seizure generation, such as the hippocampus and cortex, while being largely absent from areas like the cerebellum and brainstem, where off-target drug activity often leads to dose-limiting side effects.
The clinical development program for RAP-219 has yielded compelling results, particularly in its lead indication of drug-resistant focal onset seizures. A Phase 2a proof-of-concept trial (RAP-219-FOS-201) in a heavily treatment-experienced patient population demonstrated exceptional efficacy. The study met its primary and key secondary endpoints with high statistical significance, showing a 77.8% median reduction in clinical seizure frequency and achieving complete seizure freedom in 24% of patients over an 8-week treatment period. This level of efficacy positions RAP-219 among the most promising new antiseizure medications (ASMs) in development.
This robust efficacy is complemented by a favorable safety and tolerability profile. Across Phase 1 studies in 100 healthy volunteers and the Phase 2a trial in 30 patients, RAP-219 was generally well-tolerated. No serious adverse events were reported in the Phase 2a study, all treatment-emergent adverse events were mild or moderate in severity, and the discontinuation rate due to adverse events was low. This profile appears to validate the core scientific hypothesis that precise targeting can decouple powerful efficacy from the debilitating side effects that limit the utility of many existing ASMs.
Rapport Therapeutics is pursuing a "pipeline-in-a-product" strategy for RAP-219, with active clinical programs in bipolar disorder and a planned program in peripheral neuropathic pain. A Phase 2 trial for the acute treatment of manic episodes associated with Bipolar I Disorder is currently enrolling. The program in diabetic peripheral neuropathic pain is under a clinical hold from the U.S. Food and Drug Administration (FDA) pending the submission of additional information on trial protocol design, a matter the company is working to resolve.
Future development is focused on advancing RAP-219 into pivotal Phase 3 trials for focal epilepsy, with initiation planned for the third quarter of 2026. A key strategic initiative is the development of a long-acting injectable (LAI) formulation, which has the potential to address the significant clinical challenge of medication nonadherence in epilepsy and further differentiate RAP-219 in the market. Overall, RAP-219 represents a significant advancement in precision neuroscience, with the potential to offer a transformative treatment for patients with drug-resistant focal epilepsy and other serious CNS disorders.
Epilepsy is a chronic neurological disorder characterized by an enduring predisposition to generate epileptic seizures. Seizures are broadly classified by their onset, with focal onset seizures originating from an electrical discharge within a discrete network of neurons in one or two parts of the brain.[1] While these seizures begin locally, they can propagate to involve other brain regions, sometimes leading to secondary generalization.[1] The therapeutic landscape for epilepsy has expanded significantly over the past several decades, with nearly 30 distinct antiseizure medications (ASMs) now available.[2] Despite this armamentarium, a substantial portion of the patient population remains refractory to treatment. Approximately one-third to 40% of individuals with epilepsy suffer from drug-resistant epilepsy (DRE), defined by the failure of adequate trials of two tolerated and appropriately chosen ASM schedules to achieve sustained seizure freedom.[2]
The clinical burden of DRE is profound. Uncontrolled seizures severely impair quality of life, create barriers to social integration and employment, and significantly increase the risk of physical injury, cognitive decline, and premature mortality, including sudden unexpected death in epilepsy (SUDEP).[4] The current standard of care for patients with DRE is complex and often involves a trial-and-error approach to polypharmacy, where multiple ASMs with different mechanisms of action are combined.[2] For suitable candidates, non-pharmacological options such as resective epilepsy surgery, neurostimulation devices (e.g., Vagus Nerve Stimulation, Responsive Neurostimulation, Deep Brain Stimulation), and dietary therapies may be considered.[6] However, many patients are not candidates for these interventions or fail to achieve seizure freedom with them, underscoring the persistent and urgent need for more effective and better-tolerated pharmacological treatments.[6]
A central challenge in the development of new ASMs has been navigating the delicate balance between efficacy and tolerability. Many existing medications achieve their antiseizure effect by broadly modulating neuronal excitability, for instance, by blocking voltage-gated sodium channels or enhancing GABAergic inhibition. While effective, this widespread action can lead to a constellation of dose-limiting central nervous system (CNS) side effects. Patients frequently experience sedation, dizziness, ataxia, cognitive impairment—such as memory deficits and word-finding difficulties—and neuropsychiatric symptoms like depression and irritability.[2] These adverse effects can be as debilitating as the seizures themselves and are a primary reason for treatment discontinuation and nonadherence.[2]
Although newer generations of ASMs have generally offered improved side-effect profiles compared to their predecessors, they have not fundamentally altered the epidemiology of DRE; the proportion of patients who remain refractory to treatment has held steady at around 30-40%.[2] This therapeutic plateau suggests that simply creating variations of existing mechanisms is insufficient. A true paradigm shift requires the development of novel therapeutic strategies that can more precisely target the pathophysiological networks driving seizures while sparing the physiological networks essential for normal brain function.
In this context, Rapport Therapeutics, a clinical-stage biotechnology company, has emerged with a focus on discovering and developing small molecule precision medicines for neurological and psychiatric disorders.[10] The company's scientific foundation is built upon pioneering research into the function of Receptor Associated Proteins (RAPs). RAPs are a class of proteins that interact with neurotransmitter receptors to modulate their function, trafficking, and localization, thereby creating functional receptor diversity in different brain circuits. Rapport's RAP technology platform leverages these discoveries to design product candidates with the potential to achieve neuroanatomical specificity, a differentiated approach aimed at overcoming the limitations of conventional neurology drug discovery.[11]
The company's lead investigational asset, RAP-219, is the first product candidate to emerge from this platform. It is a small molecule engineered to selectively target a specific RAP, thereby modulating receptor function only in the discrete brain regions where that RAP is expressed.[9]
Table 1: Key Characteristics of RAP-219
| Characteristic | Description | Source(s) |
|---|---|---|
| Drug Name | RAP-219 | 9 |
| Developer | Rapport Therapeutics, Inc. | 14 |
| Drug Class | Small Molecule, Investigational Antiseizure Medication (ASM) | 9 |
| Mechanism of Action | Potential first-in-class, selective negative allosteric modulator (NAM) of TARPγ8-associated AMPA receptors | 5 |
| Highest Development Phase | Phase 2 (Completed for Focal Epilepsy; Ongoing for Bipolar Mania) | 14 |
| Primary Indication Under Review | Drug-Resistant Focal Onset Seizures | 5 |
| Other Potential Indications | Bipolar Disorder, Peripheral Neuropathic Pain | 9 |
The generation and propagation of epileptic seizures are fundamentally rooted in an imbalance between excitatory and inhibitory signaling within neuronal networks, leading to episodic, abnormal, and hypersynchronous discharges.[17] Fast excitatory synaptic transmission throughout the central nervous system is mediated predominantly by the neurotransmitter glutamate acting on its receptors. A key subtype of these, the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor, is an ionotropic glutamate receptor responsible for the majority of rapid, moment-to-moment excitatory signaling.[17]
Given their central role in excitation, AMPA receptors are directly implicated in the pathophysiology of epilepsy. Excessive activation of glutamatergic pathways via AMPA receptors is a core mechanism underlying ictogenesis.[17] This has established the AMPA receptor as a clinically validated therapeutic target for seizure control.[9] The clinical approval of perampanel, a non-selective AMPA receptor antagonist, provides definitive proof-of-concept for this approach, demonstrating that inhibiting AMPA receptor function can effectively reduce seizures in patients.[17] However, because AMPA receptors are ubiquitously expressed throughout the brain, broad, non-selective blockade can disrupt normal physiological processes, leading to significant adverse effects that limit therapeutic utility.[9]
The function of AMPA receptors in the brain is not uniform. Their properties are finely tuned by a family of auxiliary subunits known as Transmembrane AMPA Receptor Regulatory Proteins (TARPs).[18] These proteins coassemble with the core AMPA receptor subunits in the neuronal membrane and act as critical modulators. TARPs influence nearly every aspect of the receptor's life cycle and function, including its trafficking to the synapse, its subcellular localization and anchoring, and its fundamental biophysical gating properties, such as activation and desensitization kinetics.[18] The differential expression of various TARP subtypes across the brain creates a rich tapestry of AMPA receptor functional diversity, allowing for specialized signaling properties in different neuronal circuits.[21]
Among the family of TARPs, TARPγ8 possesses a unique and highly restricted expression pattern that makes it an exceptionally attractive therapeutic target.[9] While the core AMPA receptor subunits are found throughout the CNS, TARPγ8 expression is concentrated in specific forebrain regions. It is highly enriched in the hippocampus, cortex, and amygdala—areas that are frequently implicated in the generation and propagation of focal seizures.[12]
Critically, TARPγ8 expression is minimal to absent in the hindbrain, particularly the cerebellum and brain stem.[12] This anatomical distinction is of profound therapeutic importance. Unwanted pharmacological activity in the cerebellum is a well-established cause of motor side effects like ataxia and dizziness, while activity in the brain stem can lead to sedation and somnolence—all common dose-limiting toxicities of existing ASMs.[9] The selective expression of TARPγ8 therefore presents a unique opportunity to develop a precision medicine: a drug that can dampen the hyperexcitability of AMPA receptors in seizure-prone regions like the hippocampus, while leaving AMPA receptor function in areas like the cerebellum untouched. This rational design forms the central hypothesis for achieving a superior therapeutic index—delivering robust efficacy with significantly improved tolerability.
RAP-219 is a novel, potent, and selective small molecule designed to exploit the unique biology of TARPγ8.[10] It acts as a negative allosteric modulator (NAM), binding to a site at the interface between the AMPA receptor and its associated TARPγ8 protein.[18] This binding does not block the glutamate binding site directly but instead modulates the receptor's function, functionally disrupting the interaction between TARPγ8 and the AMPA receptor pore-forming subunits.[26] The result is a reduction in excitatory signaling that is restricted to only those neurons that express the TARPγ8-AMPAR complex.
Preclinical studies have provided strong support for this mechanism. In various animal models of seizures, RAP-219 demonstrated significant efficacy.[18] Importantly, this antiseizure activity was achieved without inducing the motor impairment (as assessed by the rotarod test) that is characteristic of broader-acting glutamatergic modulators. This observation in preclinical models provided the first evidence of a wide therapeutic window, suggesting that the neuroanatomical specificity could translate into a clinically meaningful separation of desired and undesired effects.[22] The success of RAP-219 in the clinic could serve to validate the entire RAP technology platform, indicating that targeting other region-specific RAPs may be a viable and highly productive strategy for developing a new generation of precision medicines for a range of CNS disorders, including chronic pain and hearing disorders.[11]
To characterize the human pharmacology of RAP-219, Rapport Therapeutics conducted a comprehensive Phase 1 program involving a total of 100 healthy volunteers.[16] This program was designed to meticulously evaluate the drug's safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD). It consisted of four distinct trials: a single ascending dose (SAD) study, two multiple ascending dose (MAD) studies, and a state-of-the-art positron emission tomography (PET) study to directly assess target engagement in the brain.[5]
The Phase 1 studies elucidated a distinct PK profile for RAP-219, characterized by slow absorption and exceptionally slow elimination.
A pivotal component of the Phase 1 program was the PET trial (RAP-219-103), an innovative study that employed a companion radiotracer to directly visualize and quantify the binding of RAP-219 to its target in the living human brain.[5] This study provided a critical de-risking step, moving the drug's mechanism from a preclinical hypothesis to a clinically confirmed reality.
The PET imaging results unequivocally validated the core mechanistic premise of RAP-219. The data confirmed that the density of TARPγ8-containing AMPA receptors is highest in the hippocampus and cerebral cortex, while being minimal in the cerebellum and brainstem.[13] This provided direct human evidence for the neuroanatomical specificity that underpins the drug's therapeutic strategy.
Furthermore, the study demonstrated that the dosing regimen selected for the Phase 2a epilepsy trial (0.75 mg daily for 5 days, followed by 1.25 mg daily) was highly effective at achieving target engagement. This regimen was shown to achieve and exceed the target receptor occupancy (RO) range of 50-70%—the level associated with maximal efficacy in preclinical seizure models—within just 5 days of initiating dosing.[5] This rapid and robust target engagement, confirmed in vivo, provided a strong pharmacodynamic rationale for the subsequent efficacy trial.
A sophisticated pharmacological insight emerged from the comparison of the SAD and MAD trial safety data, which directly informed the clinical dosing strategy. In the SAD trial, CNS-related treatment-emergent adverse events (TEAEs), such as dizziness and anxiety, were observed at the highest single doses of 2 mg and 3 mg.[22] However, in the MAD trial, these same adverse events were notably absent, even when the accumulated steady-state $C_{max}$ on Day 28 (following a daily dose of 1.25 mg) was substantially greater than the peak concentration achieved after a single 3 mg dose.[22]
This discrepancy strongly indicates that the tolerability of RAP-219 is not dictated by the absolute plasma concentration, but rather by the rate of increase in drug exposure. A rapid rise in concentration appears to trigger adverse events, whereas a slow, gradual accumulation allows the central nervous system to adapt, enabling patients to tolerate high, therapeutically effective steady-state levels. This understanding provides a clear and compelling justification for the use of a slow dose-titration schedule in the clinic (e.g., starting at 0.75 mg and increasing to 1.25 mg), a strategy designed to optimize tolerability without compromising the achievement of therapeutic target engagement.[22] This principle also lends powerful support to the strategic development of a long-acting injectable formulation, which, by its nature, would provide a very slow and steady release of the drug, potentially maximizing tolerability.
The Phase 2a clinical trial of RAP-219 in focal onset seizures, designated RAP-219-FOS-201 (NCT06377930), was a multi-center, open-label, proof-of-concept study designed to provide a robust and efficient evaluation of the drug's efficacy and safety.[10] A key innovation in the trial's design was the exclusive enrollment of patients who had been implanted with the NeuroPace RNS® System, a responsive neurostimulation device used for the treatment of drug-resistant epilepsy.[5]
The RNS System continuously monitors intracranial electroencephalography (iEEG) from electrodes placed at the seizure focus, providing a direct, real-time window into the brain's electrical activity. The device is programmed to detect and record specific patterns of epileptiform activity, including "long episodes" (LEs)—organized discharges typically exceeding 30 seconds in duration.[18] LEs are a recognized and validated objective biomarker of seizure activity, and their frequency has been shown to correlate strongly with the frequency of clinically observable seizures.[5] This trial represented the first time a novel ASM was prospectively evaluated using this objective biomarker as a primary endpoint, allowing for a highly sensitive and continuous assessment of drug effect that is less subject to the reporting biases of traditional seizure diaries.[5]
The trial enrolled 30 adult patients diagnosed with drug-resistant focal onset seizures, a population that accurately reflects the intended target for RAP-219.[5] The participants represented a particularly difficult-to-treat cohort, consistent with expectations for future pivotal trials. The median number of concomitant ASMs at baseline was three, indicating that these patients had failed multiple previous therapies. The most frequently used background medications were lamotrigine (50%), levetiracetam (40%), and cenobamate (37%), a modern and highly effective ASM, underscoring the high degree of treatment resistance in this population.[5] The mean age of participants was 40.1 years, and the mean time since their first seizure was 16.6 years, confirming the chronic and refractory nature of their condition.[5]
Table 2: Summary of Phase 2a (RAP-219-FOS-201) Trial Design and Patient Demographics
| Parameter | Description | Source(s) |
|---|---|---|
| Trial Identifier | NCT06377930 (RAP-219-FOS-201) | 5 |
| Phase | 2a Proof-of-Concept | 30 |
| Design | Multi-center, open-label | 10 |
| N | 30 | 5 |
| Patient Population | Adults (18-65) with drug-resistant focal epilepsy and an implanted RNS® System | 30 |
| Key Demographics | Mean Age: 40.1 years; Sex: 18 male, 12 female | 5 |
| Baseline Disease Severity | Median 3 concomitant ASMs | 5 |
| Treatment Regimen | 0.75 mg oral tablet daily for 5 days, then 1.25 mg daily for the remainder of the 8-week treatment period | 10 |
The RAP-219-FOS-201 trial yielded exceptionally strong and statistically robust efficacy results across both its primary biomarker endpoint and key secondary clinical endpoints.
The magnitude of these effects, particularly the 24% seizure freedom rate, is remarkable within the context of DRE clinical trials. Historically, new ASMs tested in similar add-on therapy settings often yield seizure-free rates in the low single digits.[4] The efficacy demonstrated by RAP-219 is comparable to that of cenobamate, a recently approved ASM that established a new benchmark for efficacy in DRE with a seizure-free rate of 21% in its pivotal trial.[4] This places RAP-219 in the highest tier of investigational ASMs and suggests a potential best-in-class efficacy profile. Furthermore, the strong concordance between the objective LE biomarker data and the patient-reported clinical seizure outcomes provides a powerful dual validation of the drug's potent antiseizure activity, lending high confidence to the results.
Table 3: Key Efficacy Endpoints from the Phase 2a Trial in Focal Onset Seizures
| Endpoint Category | Measure | Result | p-value | Source(s) |
|---|---|---|---|---|
| Long Episodes (LEs) - Primary | Patients with ≥30% reduction in LEs | 85.2% | <0.0001 | 5 |
| Median reduction in LE frequency | 71.0% | 0.0001 | 10 | |
| Clinical Seizures (CS) - Secondary | Median reduction in CS frequency | 77.8% | 0.01 | 10 |
| Patients with ≥50% reduction in CS | 72.0% | <0.0001 | 5 | |
| Patients achieving seizure freedom (8 weeks) | 24.0% | <0.0001 | 5 |
The safety and tolerability of RAP-219 have been evaluated in a cumulative population of 130 individuals, including 100 healthy volunteers across four Phase 1 trials and 30 patients with focal epilepsy in the Phase 2a study.[16] Across this entire clinical program, RAP-219 has demonstrated a profile of being generally safe and well-tolerated.[5]
The safety data from the RAP-219-FOS-201 trial were highly encouraging and consistent with a favorable tolerability profile, especially in the context of the profound efficacy observed.
Table 4: Summary of Treatment-Emergent Adverse Events (TEAEs) ≥10% Incidence from the Phase 2a Trial
| Adverse Event | Incidence | Number of Patients (N=30) | Source(s) |
|---|---|---|---|
| Dizziness | 26.7% | 8 | 5 |
| Headache | 16.7% | 5 | 5 |
| Fatigue | 13.3% | 4 | 5 |
| Fall | 10.0% | 3 | 5 |
| Nausea | 10.0% | 3 | 5 |
| Somnolence | 10.0% | 3 | 5 |
The overall safety profile of RAP-219 appears to be highly competitive. The combination of best-in-class level efficacy with a low discontinuation rate and a lack of any serious safety signals is a critical attribute for any new ASM seeking to gain adoption in the DRE population. More specifically, the profile provides strong clinical validation for the precision neuroscience hypothesis that underpins the drug's design.
A notable finding from the broader clinical program was the absence of sedation or motoric impairments in healthy volunteers treated with RAP-219, effects that are commonly associated with many ASMs, particularly those that broadly modulate excitatory or inhibitory systems.[13] While somnolence was reported by a small number of patients in the Phase 2a trial, the overall profile is consistent with the drug's targeted mechanism of action, which spares AMPA receptor modulation in the cerebellum and brainstem. The ability to achieve profound seizure reduction without inducing the classic signs of widespread glutamatergic blockade provides compelling evidence that the strategy of neuroanatomical targeting can successfully separate pathological network activity from normal physiological function.
Leveraging the potent and selective mechanism of RAP-219 and the favorable pharmacokinetic and safety profile established in early clinical development, Rapport Therapeutics is pursuing a "pipeline-in-a-product" strategy. This involves evaluating the compound's therapeutic potential across multiple CNS disorders where the underlying pathophysiology is believed to involve dysregulation of AMPA receptor biology.[9] In addition to focal epilepsy, the company has initiated or planned clinical programs in bipolar disorder and peripheral neuropathic pain.
The rationale for exploring RAP-219 in bipolar disorder stems from growing evidence implicating glutamatergic system hyperactivity in the pathophysiology of manic episodes. By selectively modulating AMPA receptors in key limbic circuits like the amygdala and hippocampus, RAP-219 may offer a novel approach to stabilizing mood without the broad side effects of existing treatments.
The investigation of RAP-219 for neuropathic pain is supported by the known expression of TARPγ8-containing AMPA receptors in critical pain-processing pathways, including the spinal cord dorsal horn and somatosensory cortices.[26] Modulating excitability in these circuits represents a promising strategy for pain relief.
The ambitious strategy of pursuing three distinct and high-value CNS indications simultaneously highlights the company's confidence in the broad applicability of RAP-219's mechanism. However, the clinical hold in DPNP serves as a practical reminder that each indication presents unique developmental and regulatory challenges. The translation of efficacy from epilepsy to disorders with different endpoints, such as psychiatric rating scales in mania or pain scores in neuropathy, will require carefully designed and well-justified clinical trials.
Following the highly successful outcome of the Phase 2a proof-of-concept study, Rapport Therapeutics is preparing to advance RAP-219 into the final stage of clinical development for drug-resistant focal onset seizures. The company has outlined a clear regulatory and clinical path forward. An end-of-Phase 2 meeting with the FDA is planned for the fourth quarter of 2025 to discuss the Phase 3 program design and the data package required for a New Drug Application (NDA) submission.[10]
Contingent on productive regulatory feedback, Rapport plans to initiate two pivotal Phase 3 trials in the third quarter of 2026.[5] These trials will be designed to confirm the efficacy and safety of RAP-219 in a larger patient population and will likely employ traditional, well-established regulatory endpoints for ASMs, such as the median percent reduction in seizure frequency and the proportion of patients achieving a ≥50% seizure reduction over a defined treatment period.[23]
A key element of Rapport's long-term strategy for RAP-219 is the development of a long-acting injectable (LAI) formulation.[5] This initiative is not merely a life-cycle extension but a core value driver that directly addresses a major unmet need in the management of epilepsy. Medication nonadherence is a pervasive and critical problem in this patient population, with studies indicating that up to 50% of patients are nonadherent to their prescribed oral ASM regimens.[10] This can lead to sub-therapeutic drug levels, resulting in breakthrough seizures, increased morbidity, and higher healthcare costs.
An LAI formulation of RAP-219, which would be the first potential depot ASM, could offer a transformative solution to this challenge. By providing consistent, steady drug exposure over several weeks or months from a single administration, an LAI would improve convenience, eliminate the burden of daily dosing, and, most importantly, ensure adherence. The specific PK/PD profile of RAP-219—with its long half-life and tolerability that is dependent on a slow rate of exposure increase—makes it an ideal candidate for an LAI delivery system. This formulation has the potential to significantly improve real-world effectiveness and expand the clinical utility of RAP-219 across all its potential indications.[10]
Should the positive results from Phase 2a be replicated in pivotal trials, RAP-219 is poised to become a highly competitive agent in the ASM market. Its unique value proposition rests on the combination of potentially best-in-class efficacy, comparable to the most effective modern ASMs like cenobamate, with a differentiated and potentially superior tolerability profile derived from its precision mechanism. This combination could position RAP-219 as a preferred adjunctive therapy for patients with drug-resistant focal epilepsy who have failed multiple prior treatments. Furthermore, if the "pipeline-in-a-product" strategy proves successful, with positive readouts in bipolar mania and/or peripheral neuropathic pain, the commercial ceiling for the asset would expand dramatically, establishing it as a major new therapy across multiple areas of high unmet need in neurology and psychiatry.
RAP-219 has emerged as a leading example of the promise of precision neuroscience. It is an investigational therapy founded on a rational, innovative mechanism of action that has been rigorously validated in human neuroimaging studies. The clinical data generated to date in drug-resistant focal epilepsy are exceptionally strong, demonstrating a magnitude of efficacy that is both statistically robust and profoundly meaningful for patients. This high level of efficacy is paired with a favorable safety and tolerability profile that appears to be the direct result of the drug's neuroanatomical specificity.
The path forward is defined by several key catalysts: the successful execution of the pivotal Phase 3 program in epilepsy, the data readout from the Phase 2 trial in bipolar mania, and the resolution of the clinical hold for the neuropathic pain program. The parallel development of a novel LAI formulation represents a significant opportunity to further enhance the drug's clinical and commercial profile. In sum, RAP-219 stands as a compelling asset with the potential to become a transformative treatment for patients suffering from focal epilepsy and other serious disorders of the central nervous system.
Published at: October 27, 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.