Vixotrigine (DB11706), also known by its development codes BIIB074, CNV1014802, and GSK-1014802, is an investigational, orally administered small molecule developed for the treatment of chronic neuropathic pain. Originating at GlaxoSmithKline and advanced through Convergence Pharmaceuticals before its acquisition by Biogen, Vixotrigine was engineered to address the significant tolerability limitations of existing voltage-gated sodium channel (Nav) blockers, such as carbamazepine. Its core pharmacological feature is a state- and use-dependent mechanism of action, designed to selectively inhibit pathologically hyperexcitable neurons characteristic of pain states while sparing normal physiological nerve signaling. This approach was intended to deliver targeted analgesia with a superior safety profile.
From a physicochemical and pharmacokinetic standpoint, Vixotrigine was an exemplary drug candidate. It possesses favorable "drug-like" properties, adheres to Lipinski's Rule of Five, and exhibits a predictable pharmacokinetic profile with rapid absorption, a half-life of approximately 11 hours suitable for twice-daily dosing, and linear kinetics without the autoinduction that complicates the use of older agents. Clinical studies consistently affirmed its primary strategic goal: Vixotrigine was generally well-tolerated across a range of doses, with a manageable side-effect profile consisting primarily of mild-to-moderate, transient central nervous system effects like dizziness and headache.
Despite this success in achieving its safety and tolerability objectives, Vixotrigine's clinical development was ultimately terminated due to a persistent and irreconcilable pattern of inconsistent efficacy. While an early Phase IIa study in trigeminal neuralgia (TN) showed highly promising and statistically significant pain reduction, this momentum could not be sustained. A program in lumbosacral radiculopathy was halted after a larger Phase IIb study failed to replicate positive initial findings. The subsequent Phase III program in TN was withdrawn by Biogen before completion, signaling a loss of confidence in the drug's lead indication.
The decisive blow to the program came from the Phase II CONVEY study in painful small fiber neuropathy (SFN). The trial yielded a paradoxical and pharmacologically incoherent result: the lower 200 mg twice-daily dose met its primary endpoint for pain reduction, while the higher 350 mg twice-daily dose failed to do so. This lack of a clear dose-response relationship created insurmountable uncertainty, rendering the design of a viable Phase III program exceptionally risky and strategically untenable. Faced with a cumulative history of unreproducible efficacy signals across multiple indications, Biogen officially discontinued all development of Vixotrigine in late 2022. The story of Vixotrigine serves as a critical case study in modern drug development, illustrating that even a molecule with an elegant mechanism, favorable pharmacokinetics, and a successful safety profile cannot overcome the absolute requirement for robust, consistent, and clinically meaningful efficacy.
Neuropathic pain represents a substantial global health burden, affecting an estimated 6.9% to 10% of the general population.[1] It is a complex, chronic pain state caused by a lesion or disease of the somatosensory nervous system. Unlike nociceptive pain, which arises from tissue damage, neuropathic pain is pathological and often persists long after the initial injury has healed. This condition leads to a severe degradation in quality of life, frequently accompanied by comorbidities such as anxiety, depression, cognitive impairment, and significant sleep disturbance.[1]
The clinical development of Vixotrigine targeted several specific and severe forms of neuropathic pain where existing treatments are inadequate. Trigeminal neuralgia (TN), a lead indication for the drug, is an archetypal example. It is characterized by sudden, severe, and debilitating episodes of facial pain, often described as electric shock-like paroxysms, which can be triggered by trivial stimuli.[2] Similarly, small fiber neuropathy (SFN) involves damage to small peripheral nerve fibers, resulting in severe burning or tingling pain, typically in the hands and feet. SFN is frequently associated with diabetes mellitus and, like many neuropathic conditions, lacks effective, curative therapies, leaving patients with a significant unmet medical need.[5]
The therapeutic landscape that Vixotrigine was designed to enter is dominated by older medications with significant drawbacks. The first-line pharmacological treatments for conditions like TN are voltage-gated sodium channel blockers, primarily carbamazepine and its analogue, oxcarbazepine.[9] These drugs function by non-selectively blocking Nav channels, thereby reducing neuronal excitability.
While their efficacy in reducing neuropathic pain is established, their clinical utility is severely compromised by a poor tolerability profile. Patients frequently experience central nervous system (CNS) side effects, including cognitive impairment, tiredness, memory problems, dizziness, and ataxia.[2] Furthermore, these agents are associated with a risk of serious and potentially life-threatening adverse events, such as hyponatremia, decreased hematologic cell counts, and severe cutaneous reactions like Stevens-Johnson syndrome.[3]
The use of these drugs is further complicated by unfavorable pharmacokinetic properties. Carbamazepine is a potent inducer of its own metabolism (autoinduction) and other hepatic enzymes, leading to numerous drug-drug interactions and requiring a slow, careful dose titration over several weeks to reach a therapeutic level and manage side effects.[2] Over time, many patients also develop tachyphylaxis, a phenomenon where the drug's efficacy diminishes.[2] This combination of poor tolerability, complex dosing, and risk of serious side effects created a clear and compelling clinical need for a novel sodium channel blocker that could offer comparable or superior efficacy with an improved safety and convenience profile.[2]
Vixotrigine was developed to directly address the shortcomings of the existing standard of care. Originally developed by GlaxoSmithKline and later advanced by Convergence Pharmaceuticals (which was subsequently acquired by Biogen), Vixotrigine is a novel, orally administered small molecule analgesic.[11]
The core of its innovative design lies in its distinct mechanism of action as a voltage- and use-dependent sodium channel blocker.[2] The central hypothesis behind its development was that this state-dependent mechanism would confer selectivity. By preferentially binding to Nav channels in their inactivated state—a state more prevalent in neurons undergoing the rapid, high-frequency firing characteristic of pathological pain signaling—Vixotrigine was expected to selectively dampen hyperexcitability in pain pathways. Simultaneously, it was hypothesized to have a minimal effect on neurons firing at normal, low physiological frequencies, such as those involved in cardiac function and normal CNS activity.[10] This targeted approach was designed to achieve a wider therapeutic window, delivering potent analgesia with a significantly improved safety and tolerability profile compared to older, non-selective agents.[10]
This "safety-first" development strategy was a logical response to the market need. The primary goal was not necessarily to demonstrate overwhelmingly superior efficacy to carbamazepine, but rather to offer a much better-tolerated alternative that was easier for clinicians to prescribe and for patients to take, notably without the need for lengthy titration.[2] However, this strategic positioning placed a critical burden on the drug: it had to demonstrate, at a minimum, robust and consistent efficacy. A drug that is only marginally safer but less effective than the standard of care holds little clinical value. This fundamental requirement for clear efficacy would ultimately prove to be the program's greatest challenge and foreshadow its eventual discontinuation. The development trajectory of Vixotrigine demonstrates a common and difficult challenge in creating "next-generation" therapies: an improved safety profile, while highly desirable, cannot compensate for an efficacy signal that is weak, inconsistent, or unreproducible.
Vixotrigine is a small molecule drug that has been identified by several names and development codes throughout its history, reflecting its passage through different pharmaceutical companies.
Vixotrigine's chemical structure is defined by a pyrrolidine core with specific stereochemistry, linked to substituted aromatic rings.
An analysis of Vixotrigine's computed physicochemical properties reveals a molecule well-optimized for development as an oral therapeutic. It belongs to the chemical class of pyrrolidines, and more specifically, is a derivative of the amino acid proline.[11] Its properties align closely with established guidelines for oral bioavailability and central nervous system penetration, such as Lipinski's Rule of Five, for which no violations are reported.[22]
Collectively, these excellent physicochemical properties supported the drug's development strategy, predicting that it could be effectively administered orally and would achieve sufficient exposure in both the peripheral and central nervous systems to engage its targets.[10]
Table 1: Key Chemical Identifiers and Physicochemical Properties of Vixotrigine
Property | Value | Source(s) |
---|---|---|
Common Name | Vixotrigine | 16 |
Synonyms | Raxatrigine, BIIB074, CNV1014802, GSK-1014802 | 11 |
DrugBank ID | DB11706 | 17 |
CAS Number | 934240-30-9 (free base) | 16 |
IUPAC Name | (2S,5R)-5-[(2-fluorophenyl)methoxy]phenyl]pyrrolidine-2-carboxamide | 16 |
Molecular Formula | 16 | |
Molecular Weight | 314.36 g/mol | 16 |
LogP (AlogP) | 2.68 | 23 |
TPSA | 64.35 | 22 |
Rotatable Bonds | 5 | 17 |
H-Bond Donors (Lipinski) | 3 | 23 |
H-Bond Acceptors (Lipinski) | 4 | 23 |
Lipinski's Ro5 Violations | 0 | 22 |
The fundamental mechanism of action for Vixotrigine is the blockade of voltage-gated sodium channels (Navs).[15] These ion channels are critical for the initiation and propagation of action potentials in excitable cells, including neurons. The key innovation in Vixotrigine's design is its
state- and use-dependent mode of inhibition.[2] This means the drug exhibits a higher affinity for Nav channels that are in an open or inactivated state compared to those in a resting state.
This state-dependent binding is the pharmacological basis for its intended therapeutic selectivity. In pathological conditions like neuropathic pain, nociceptive (pain-sensing) neurons often display ectopic, high-frequency firing. This sustained activity causes a greater proportion of Nav channels to occupy the open and inactivated states, creating a preferential target for Vixotrigine.[10] By binding to and stabilizing the inactivated state of the channel, Vixotrigine prolongs the recovery period (repriming) before the channel can open again, effectively dampening the excessive neuronal firing that underlies the sensation of pain.[26] Conversely, in healthy tissues where neurons are firing at normal, low physiological frequencies, most Nav channels are in the resting state, for which Vixotrigine has low affinity. This was the central hypothesis for achieving an improved therapeutic index, maximizing analgesic efficacy while minimizing side effects associated with blocking normal neuronal function.[10]
The description of Vixotrigine's specific molecular targets evolved significantly over the course of its development, a progression that has important strategic implications. There are nine functional mammalian Nav channel subtypes (Nav1.1–Nav1.9), each with distinct tissue distributions and physiological roles.
This "target drift" from a highly selective, novel agent to a broad-spectrum blocker represents more than a simple scientific reclassification. It fundamentally altered the drug's strategic narrative and value proposition. The initial positioning as a selective Nav1.7 inhibitor would have generated immense interest, suggesting a precision-medicine approach with the potential to revolutionize pain treatment. The final characterization as a broad-spectrum blocker, even with the advantage of state-dependency, repositioned Vixotrigine as an incrementally improved version of existing drugs like carbamazepine. Its story changed from one of "novel mechanism" to "better-tolerated version of an old mechanism." This is a far less compelling narrative for regulators, clinicians, and investors, and it raises the bar for demonstrating a truly superior clinical profile. The failure to deliver on the promise of high target selectivity may have contributed to the heightened scrutiny of its mixed efficacy results in later clinical trials.
When compared directly to the established first-line therapy, carbamazepine, preclinical studies highlighted Vixotrigine's potential advantages. While both drugs act as broad-spectrum Nav channel blockers, Vixotrigine was shown to have higher potency and a more profound state-dependent inhibitory effect.[1] This superior state-dependency was a key preclinical finding that supported the hypothesis that Vixotrigine could achieve a better separation between efficacy and adverse effects.
In addition to its primary activity as a Nav channel blocker, Vixotrigine was also identified as a potent inhibitor of monoamine oxidase B (MAO-B), with a reported of approximately 4 nM.[22] MAO-B is an enzyme involved in the metabolism of neurotransmitters like dopamine. While MAO-B inhibitors are used in the treatment of Parkinson's disease, the clinical relevance of this secondary activity to Vixotrigine's potential analgesic effect was not fully explored in the available documentation. Nonetheless, it represents an additional pharmacological property that could contribute to its overall biological effects.
The pharmacokinetic (PK) profile of Vixotrigine in humans has been well-characterized through Phase I studies in healthy volunteers. The results show a molecule with properties conducive to a straightforward oral dosing regimen, a key advantage over older anticonvulsants used in neuropathic pain.
Following oral administration, Vixotrigine is rapidly and extensively absorbed.[1] Peak plasma concentrations (
) are consistently achieved within 1 to 2 hours post-dose across a wide range of single doses, from 75 mg to 825 mg.[1] This rapid absorption allows for a quick onset of systemic exposure.
Preclinical studies in multiple species indicated that Vixotrigine undergoes moderate to extensive tissue distribution, with a volume of distribution at steady state () that exceeded total body water.[10] This finding was confirmed in human studies, where the estimated volume of distribution was approximately 262 L.[29] This extensive distribution is consistent with a molecule designed to act on targets in both the central and peripheral nervous systems.
The primary route of metabolism for Vixotrigine is glucuronidation, a Phase II metabolic reaction catalyzed by the uridine diphosphate-glucuronosyltransferase (UGT) family of enzymes.[28] This metabolic pathway has implications for potential drug-drug interactions. A study investigating the co-administration of Vixotrigine with carbamazepine, a known inducer of several UGT enzymes, demonstrated a clinically relevant interaction. Carbamazepine administration led to a 31.6% decrease in the area under the curve (AUC) and a 26.3% decrease in the maximum concentration (
) of Vixotrigine. While this effect was characterized as potentially "clinically unimportant," it highlights a metabolic vulnerability that could require dose adjustments in a real-world clinical setting.[28]
While human excretion data is limited in the provided materials, preclinical studies in rats provide insight into the elimination pathways. Following a single oral dose of radiolabeled Vixotrigine, the drug and its metabolites were eliminated through both urine (approximately 58% of the dose) and feces (approximately 36%).[9] Elimination in this species was rapid, with nearly 93% of the administered dose recovered within 24 hours.[9]
The terminal elimination half-life () of Vixotrigine in humans is consistently reported to be in the range of 9-13 hours, with a mean of approximately 11 hours.[1] This pharmacokinetic parameter is ideal for supporting a convenient
twice-daily (BID) dosing regimen, which was used in most of the Phase II clinical trials.[10]
Upon repeat dosing, steady-state plasma concentrations are achieved by approximately day 5.[9] As expected from its half-life, a twofold increase in drug accumulation was observed with BID dosing compared to once-daily (QD) administration.[9] Importantly, Vixotrigine exhibits linear pharmacokinetics, meaning that its exposure (AUC and
) increases proportionally with the dose. This predictable behavior, along with a lack of evidence for autoinduction, stands in stark contrast to the complex, non-linear, and autoinducing PK of carbamazepine.[29]
This "clean" and predictable pharmacokinetic profile was a cornerstone of Vixotrigine's development strategy. The simplicity of a BID dosing schedule without the need for slow, complex titration was a major intended advantage over the standard of care. This PK profile was a key enabler of the "improved tolerability and convenience" value proposition. However, this advantage is only meaningful if the drug's efficacy is robust and reliable. The straightforward PK of Vixotrigine meant that the subsequent efficacy failures in clinical trials could not be easily attributed to complex dosing or unpredictable drug exposure, making the inconsistent results even more difficult to explain and defend.
Table 2: Summary of Vixotrigine Pharmacokinetic Parameters in Humans
Parameter | Value | Source(s) |
---|---|---|
(Time to Peak Concentration) | ~1–2 hours | 1 |
(Terminal Half-life) | ~11 hours | 1 |
Dosing Regimen | Twice-daily (BID) | 10 |
Time to Steady State | ~5 days | 9 |
Primary Metabolism | UGT Glucuronidation | 28 |
Key Differentiator | Linear kinetics, no autoinduction | 29 |
Vixotrigine underwent a broad clinical development program, exploring its utility across several neuropathic pain conditions and, briefly, in psychiatric disorders. The program advanced to Phase III for its lead indication before its eventual termination.[16]
Trigeminal neuralgia was the most advanced and, initially, the most promising indication for Vixotrigine.
The development in SFN culminated in the Phase II CONVEY study (NCT03339336), another EERW trial that would ultimately deliver the most confounding results of the entire program.[6]
This paradoxical result from the CONVEY study was likely the decisive factor in the drug's demise. A result where a lower dose is effective but a higher dose is not defies a logical dose-response relationship. This is not merely a "failed" trial but an incoherent one, creating profound uncertainty about the drug's true efficacy and optimal dose. It would be nearly impossible to design a successful Phase III program based on such contradictory data, as regulators would demand a clear and understandable dose-response. This outcome effectively rendered the path forward for Vixotrigine in SFN strategically untenable.
The clinical development in PLSR followed a pattern that would become characteristic of the Vixotrigine program: an encouraging early signal that could not be replicated in subsequent, more rigorous testing.
This pattern of unreproducible efficacy, seen first in PLSR and later echoed by the withdrawal of the TN program and the paradoxical CONVEY results, points to a fundamental issue with the drug's analgesic effect. It suggests that Vixotrigine's efficacy signal was likely small and not robust enough to consistently and reliably separate from the high placebo response rates that are characteristic of chronic pain trials.[44]
Table 3: Overview of Key Clinical Trials for Vixotrigine
Indication | Trial ID(s) | Phase | Status | Key Outcome |
---|---|---|---|---|
Trigeminal Neuralgia | 1014802/202 (PoC) | IIa | Completed | Positive: Statistically significant reduction in pain paroxysms and daily pain score. |
Trigeminal Neuralgia | NCT03070132, NCT03637387 | III | Withdrawn | Negative: Discontinued by sponsor before completion. |
Small Fiber Neuropathy | NCT03339336 (CONVEY) | II | Completed | Paradoxical: 200mg dose met primary endpoint; 350mg dose failed. |
Lumbosacral Radiculopathy | NCT01561027 | II | Completed | Positive: Initial statistically significant improvement in neuropathic pain. |
Lumbosacral Radiculopathy | NCT02935608 | IIb | Completed | Negative: Failed to confirm efficacy; indication discontinued. |
Bipolar Disorder | N/A | I | Discontinued | Negative: Development halted in early phase. |
A consistent finding across the entire Vixotrigine clinical development program was its favorable safety and tolerability profile. In numerous Phase I studies in healthy volunteers and Phase II studies in various patient populations, the drug was consistently described as "generally well tolerated".[2]
Phase I single ascending dose (SAD) and multiple ascending dose (MAD) studies established that oral Vixotrigine was well-tolerated at single doses up to 825 mg and at multiple doses up to 450 mg twice daily.[9] This positive safety profile remained consistent throughout later-stage trials, with no new or unexpected safety signals emerging.[42] Furthermore, studies found no evidence of abuse potential, an important consideration for a centrally-acting analgesic.[14]
The adverse event profile of Vixotrigine was predictable for a centrally-acting sodium channel blocker. The majority of reported AEs were mild to moderate in severity and often transient.[39]
Serious adverse events (SAEs) were infrequent in the Vixotrigine clinical program. Discontinuation rates due to AEs were generally low, further supporting the drug's good tolerability. For instance, during the 4-week open-label period of the CONVEY study where all 265 participants received Vixotrigine 350 mg BID, only 5.3% of patients discontinued treatment due to an adverse event.[42] In the subsequent 12-week double-blind portion of that study, the rate of discontinuation due to AEs was notably lower in the Vixotrigine arms (5.0% for 200 mg BID and 0% for 350 mg BID) than in the placebo arm (7.3%).[41]
The success of Vixotrigine in meeting its primary design goal of being a well-tolerated sodium channel blocker presents a tragic irony. The drug effectively solved the tolerability problem that plagues older agents like carbamazepine. It was not a drug that failed because it was unsafe; it was a drug that failed despite being safe. This outcome underscores a fundamental principle of modern drug development: safety is a necessary, but not sufficient, condition for success. An excellent safety profile cannot rescue a drug from a weak or inconsistent efficacy profile. Vixotrigine's story is a poignant cautionary tale for development strategies that prioritize improving the safety of an established drug class without ensuring the new agent's efficacy is, at a minimum, robust and non-inferior.
Table 4: Summary of Common Adverse Events (≥5% Incidence) in the CONVEY Study (Double-Blind Period)
Adverse Event | Placebo (N=41) | Vixotrigine 200mg BID (N=40) | Vixotrigine 350mg BID (N=41) |
---|---|---|---|
Falls | 4.9% | 10.0% | 7.3% |
Nasopharyngitis | 7.3% | 7.5% | 2.4% |
Muscle Spasm | 2.4% | 5.0% | 2.4% |
Urinary Tract Infection | 0% | 5.0% | 4.9% |
Dizziness | 4.9% | 2.5% | 4.9% |
Headache | 4.9% | 2.5% | 7.3% |
Note: Data derived from narrative descriptions of the CONVEY study results. Precise percentages may vary slightly based on final study reports. 39 |
The termination of the Vixotrigine development program was not a single event but a gradual process of deprioritization driven by accumulating negative or ambiguous clinical data.
The decision to terminate the Vixotrigine program was a strategic one, rooted in a comprehensive assessment of the drug's declining probability of clinical and commercial success. It was not a decision driven by safety concerns.
The primary driver was the cumulative weight of inconsistent and failed efficacy data. The pattern of an initial promising result followed by failure in a larger, confirmatory study (as seen in PLSR) and the withdrawal of the most advanced program (in TN) severely eroded confidence in the drug's fundamental analgesic effect.[11]
The paradoxical dose-response from the CONVEY study was the final, insurmountable hurdle. This scientifically incoherent result created an unacceptable level of uncertainty for any potential Phase III program, making the enormous financial investment required for such trials impossible to justify.[8]
This decision was also made within the context of a broader portfolio review at Biogen. The company was strategically shifting its resources toward assets perceived to have a higher probability of success and greater commercial potential, such as its programs in Alzheimer's disease (lecanemab) and depression (zuranolone).[47] The termination of the Vixotrigine program was a calculated business decision to cut losses on an increasingly risky asset and reallocate capital to more promising areas.
The discontinuation of the Vixotrigine program had a significant and mixed impact on Biogen's financial statements for the fourth quarter of 2022.
This financial accounting clearly illustrates that the end of the Vixotrigine program was a strategic and financial decision. The accumulating negative clinical data led to a reassessment of the asset's risk-adjusted net present value (rNPV). Once the rNPV fell below a critical threshold, the program was terminated, triggering the necessary accounting adjustments. It is a stark example of how clinical trial outcomes directly translate into corporate financial strategy and portfolio management decisions.
The trajectory of Vixotrigine, from a promising next-generation analgesic to a discontinued asset, offers critical lessons on the complexities and challenges of modern pharmaceutical development, particularly in the difficult therapeutic area of neuropathic pain.
Vixotrigine's Scientific Legacy: From a drug design perspective, Vixotrigine can be considered a success. It is a well-crafted molecule with excellent physicochemical properties and a clean, predictable pharmacokinetic profile that represented a clear improvement over older agents. It successfully validated the scientific hypothesis that a state- and use-dependent mechanism of sodium channel blockade could lead to significantly better tolerability than non-selective blockade. The drug consistently demonstrated a favorable safety profile in clinical trials, achieving the primary objective of its "safety-first" design strategy.
The Efficacy Hurdle in Neuropathic Pain: Despite its scientific elegance and good safety, Vixotrigine's downfall was its failure to clear the high bar for efficacy in neuropathic pain. Its clinical program was plagued by a pattern of promising early signals that could not be reliably reproduced in larger, more rigorous confirmatory trials. This narrative of unreproducible results, culminating in the pharmacologically incoherent outcome of the CONVEY study, ultimately sealed its fate. The story of Vixotrigine is a powerful case study on the immense difficulty of developing drugs for chronic pain, a field notorious for high placebo response rates, subjective endpoints, and patient heterogeneity, which collectively conspire to obscure a true drug effect.[44]
Final Verdict: Vixotrigine represents a strategic failure born from scientific uncertainty. It was not a dangerous or poorly designed drug; on the contrary, it was a well-engineered molecule that achieved its primary safety goals. However, its clinical effect on pain was too inconsistent, too unpredictable, and ultimately too uncertain to warrant the massive financial and operational investment of a global Phase III development program. Its history serves as a critical reminder of several fundamental truths in drug development:
Ultimately, Vixotrigine will be remembered as a cautionary tale—a scientifically sound concept and a well-behaved molecule that was defeated by the inherent biological complexity and clinical trial challenges of treating chronic neuropathic pain.
Published at: October 3, 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.