MedPath

Edivoxetine Advanced Drug Monograph

Published:Sep 7, 2025

Generic Name

Edivoxetine

Drug Type

Small Molecule

Chemical Formula

C18H26FNO4

CAS Number

1194508-25-2

Edivoxetine (LY-2216684): A Comprehensive Monograph on a Selective Norepinephrine Reuptake Inhibitor – From Pharmacological Promise to Clinical Discontinuation

Executive Summary

Edivoxetine, also known by its development code LY-2216684, is a small molecule drug developed by Eli Lilly and Company as a potent and highly selective norepinephrine reuptake inhibitor (NRI).[1] Its primary mechanism of action is the specific blockade of the norepinephrine transporter (NET), leading to increased synaptic concentrations of norepinephrine.[3] This mechanism formed the therapeutic rationale for its investigation in two major psychiatric conditions: as an adjunctive treatment for Major Depressive Disorder (MDD) in patients with a partial response to selective serotonin reuptake inhibitors (SSRIs), and as a monotherapy for Attention-Deficit/Hyperactivity Disorder (ADHD).[1]

Despite a promising pharmacological profile and evidence of target engagement in humans, the clinical development program for Edivoxetine ultimately met with definitive failure. The extensive Phase III program for MDD, comprising three large, randomized, placebo-controlled trials, failed to demonstrate that Edivoxetine, when added to an existing SSRI, provided any statistically significant benefit over adding a placebo.[5] This lack of efficacy was observed despite the drug demonstrating a clear biological effect, as evidenced by its characteristic noradrenergic safety profile.

The safety and tolerability profile of Edivoxetine was consistent with its mechanism of action, characterized by statistically significant increases in blood pressure and heart rate, as well as common noradrenergic side effects such as hyperhidrosis, nausea, and dry mouth.[7] While these effects were generally manageable, they underscored that the drug was pharmacologically active.

The primary reason for the discontinuation of its development for MDD in December 2013 was the conclusive lack of clinical efficacy.[5] This outcome was significantly influenced by an unusually high placebo response rate in the pivotal trials, a persistent challenge in CNS drug development that may have masked a modest drug effect.[9] While initial studies in pediatric ADHD showed some evidence of efficacy, a more recent and comprehensive network meta-analysis has since concluded that Edivoxetine is no more effective than placebo for ADHD, casting doubt on its viability for that indication as well.[4]

The trajectory of Edivoxetine serves as a critical case study in modern psychopharmacology. It demonstrates the profound disconnect that can exist between a well-designed molecule, a plausible neurochemical hypothesis, and meaningful clinical outcomes. Its failure challenges the therapeutic utility of highly selective noradrenergic modulation as a standalone or adjunctive strategy for MDD and highlights the formidable obstacles, particularly the placebo effect and complex trial design, that confront the development of novel antidepressants.

Molecular Profile and Mechanism of Action

The foundation of any pharmacological agent lies in its distinct molecular structure and its specific interaction with biological targets. Edivoxetine was engineered as a precise chemical entity designed to selectively modulate the noradrenergic system with high fidelity.

Chemical Identity and Physicochemical Properties

Edivoxetine is a synthetic small molecule belonging to the morpholine and phenylethyl alcohol chemical classes.[11] Its development as a single, specific stereoisomer reflects a sophisticated approach to drug design aimed at maximizing potency and minimizing off-target effects. The synthesis process utilizes D-serine to establish the molecule's chirality, ensuring the production of the desired enantiomer.[13] This level of chemical precision is critical, as the therapeutic activity of many psychoactive compounds is highly dependent on their three-dimensional structure. The key chemical and regulatory identifiers for Edivoxetine are consolidated in Table 1.

The molecule's physicochemical properties, such as a LogP of 1.851 and a polar surface area of 59.95 Ų, are consistent with those of a centrally-acting oral drug, suggesting good potential for absorption and blood-brain barrier penetration.[14]

Table 1: Key Chemical and Regulatory Identifiers for Edivoxetine

ParameterValueSource(s)
Generic NameEdivoxetine1
DrugBank IDDB091841
Code Name(s)LY-2216684, LY2216684, EDP1251
TypeSmall Molecule1
CAS Number (Base)1194508-25-22
CAS Number (HCl)1194374-05-42
Molecular FormulaC18​H26​FNO4​1
Average Weight339.407 g/mol1
Monoisotopic Weight339.184586484 g/mol1
IUPAC Name(1R)-2-(5-fluoro-2-methoxyphenyl)-1--1-(tetrahydro-2H-pyran-4-yl)ethanol2
SMILESCOC1=C(C=C(C=C1)F)C[C@]([C@@H]2CNCCO2)(C3CCOCC3)O2
InChIKeyCPBHSHYQQLFAPW-ZWKOTPCHSA-N2

Primary Pharmacology: Selective Inhibition of the Norepinephrine Transporter (NET)

The primary mechanism of action of Edivoxetine is the potent and selective inhibition of the presynaptic norepinephrine transporter (NET).[3] The NET is a protein responsible for the reuptake of norepinephrine from the synaptic cleft back into the presynaptic neuron, thereby terminating its signaling activity.[3] By blocking this transporter, Edivoxetine effectively increases the concentration and prolongs the residence time of norepinephrine in the synapse, enhancing noradrenergic neurotransmission.[3]

This targeted action was the cornerstone of its development strategy. The "selective NRI" hypothesis posits that enhancing noradrenergic function can specifically alleviate symptoms of depression and ADHD that are often refractory to purely serotonergic agents, such as fatigue, anergia, apathy, and deficits in executive function and cognitive control.[3] The deliberate selectivity of Edivoxetine for the NET was designed to avoid interactions with other monoamine transporters, such as the serotonin transporter (SERT) and the dopamine transporter (DAT), as well as other neuroreceptors.[3] This high degree of selectivity was intended to produce a "cleaner" pharmacological profile, theoretically minimizing side effects commonly associated with less selective antidepressants, such as the sexual dysfunction, weight gain, and gastrointestinal disturbances often seen with SSRIs and older tricyclic agents.[3]

While specific binding affinity constants (e.g., Ki​ values) for Edivoxetine at the human NET, SERT, and DAT are not detailed in the available documentation, its consistent description as "highly selective" implies a binding affinity for NET that is orders of magnitude greater than for SERT or DAT.[3] This profile distinguishes it from serotonin-norepinephrine reuptake inhibitors (SNRIs) like venlafaxine or duloxetine, which potently inhibit both NET and SERT, and from agents like bupropion, which primarily inhibits NET and DAT. The clinical failure of Edivoxetine in MDD, therefore, represents a significant challenge to the hypothesis that a highly selective increase in noradrenergic tone is a sufficient strategy to produce a robust antidepressant effect in patients who have not fully responded to serotonergic agents.

Pharmacokinetic and Pharmacodynamic Profile

A drug's clinical utility is governed by its pharmacokinetics (what the body does to the drug) and its pharmacodynamics (what the drug does to the body). For Edivoxetine, studies revealed a predictable pharmacokinetic profile and clear evidence of pharmacodynamic target engagement, creating a perplexing disconnect with its ultimate lack of clinical efficacy in MDD.

Absorption, Distribution, Metabolism, and Excretion (ADME)

Edivoxetine was developed for oral administration and demonstrated pharmacokinetic properties suitable for a once-daily dosing regimen.[11]

Absorption: Following oral administration, Edivoxetine is readily absorbed, with the time to reach maximum plasma concentration (Tmax​) occurring consistently at approximately 2 hours post-dose in both pediatric and adult populations.[11]

Distribution: The drug is distributed throughout the body, and while specific plasma protein binding data is not provided, pharmacokinetic modeling in pediatric studies accounted for the apparent volume of distribution at steady state (Vss​/F), indicating standard distribution characteristics for a CNS-active compound.[11]

Metabolism: Edivoxetine undergoes extensive hepatic metabolism, primarily mediated by the cytochrome P450 (CYP) enzyme system. The isoenzymes CYP2D6 and CYP3A4 have been identified as playing the most prominent roles in its clearance.[22] This metabolic pathway is of high clinical significance. The gene encoding the CYP2D6 enzyme is highly polymorphic in the human population, leading to distinct phenotypes of poor, intermediate, extensive, and ultra-rapid metabolizers. This genetic variability can result in substantial inter-individual differences in drug exposure, which may lead to toxicity in poor metabolizers or lack of efficacy in ultra-rapid metabolizers. Such variability, if not accounted for in clinical trial design and analysis, can introduce significant "noise" into the data, potentially obscuring a true drug effect and contributing to a failure to demonstrate efficacy.

Excretion: After reaching its peak concentration, the plasma level of Edivoxetine follows a mono-exponential decline. The terminal elimination half-life (t1/2​) is consistently reported to be in the range of 4 to 6 hours.[11] This relatively short half-life is sufficient to support once-daily dosing to maintain steady-state concentrations but also suggests that missed doses could lead to rapid decreases in plasma levels, potentially increasing the risk for discontinuation symptoms.

Pharmacokinetic parameters were found to be largely comparable between children and adolescents, simplifying dosing considerations in pediatric populations.[11] Furthermore, dedicated studies were conducted to assess the pharmacokinetics in patients with renal and hepatic impairment, a standard part of drug development to establish dosing guidelines for special populations.[16]

Pharmacodynamics and Biomarker Analysis

A critical component of modern drug development is demonstrating that an investigational compound engages its intended biological target in humans at clinically relevant doses. For Edivoxetine, this was achieved through the use of a key pharmacodynamic biomarker: 3,4-dihydroxyphenylglycol (DHPG).[11]

DHPG is an intraneuronal metabolite of norepinephrine. Its concentration in plasma is dependent on the reuptake of norepinephrine into the presynaptic neuron via the NET. By inhibiting NET, Edivoxetine blocks this reuptake process, thereby reducing the amount of norepinephrine available for metabolism into DHPG. Consequently, a reduction in plasma DHPG levels serves as a direct, functional measure of NET inhibition in the central and peripheral nervous systems.[11]

Clinical studies in adolescent patients with ADHD provided clear evidence of this target engagement. Administration of Edivoxetine resulted in a maximum decrease in plasma DHPG concentrations from baseline of approximately 28%, with the most notable reduction occurring within 8 hours post-dose.[11] This finding was crucial, as it confirmed that Edivoxetine was reaching its target and exerting its intended biological effect at the doses being tested in efficacy trials.

The establishment of this relationship between drug exposure (pharmacokinetics) and biological effect (pharmacodynamics) was used to inform dose selection and guide the development of population PK/PD models.[11] However, this successful demonstration of target engagement stands in stark contrast to the drug's ultimate clinical failure in MDD. This "PK/PD disconnect" is a central element of the Edivoxetine story. It proves that successfully hitting a specific molecular target is not, by itself, sufficient to guarantee clinical efficacy in a complex neuropsychiatric disorder. The drug was performing its designed function at a biological level, but this biological action did not translate into a therapeutic effect that could be reliably distinguished from placebo.

The Clinical Development Trajectory: A Tale of Two Indications

The clinical development of Edivoxetine was pursued with significant investment, focusing primarily on MDD as an adjunctive therapy and secondarily on ADHD as a non-stimulant monotherapy. The divergent outcomes and subsequent re-evaluation of its efficacy in both indications provide a compelling narrative of the challenges inherent in CNS drug development.

The Pursuit of an Adjunctive Antidepressant for Major Depressive Disorder (MDD)

The strategic positioning of Edivoxetine in MDD was based on a strong clinical and scientific rationale. A substantial portion of patients treated for MDD with first-line SSRIs fail to achieve full remission, experiencing residual symptoms that impair functioning and increase the risk of relapse.[5] One prominent hypothesis to explain this partial response is that purely serotonergic modulation is insufficient to address the full spectrum of depressive symptoms. Augmenting an SSRI with an agent that enhances noradrenergic neurotransmission was theorized to provide a synergistic effect, potentially improving outcomes in this treatment-resistant population.[3]

Edivoxetine, as a potent and highly selective NRI, was the ideal tool to test this hypothesis. Eli Lilly launched a robust Phase III program in 2010 specifically designed to assess the benefits of Edivoxetine as an add-on therapy for patients who had achieved only a partial response to at least six weeks of stable SSRI treatment.[5]

A Critical Analysis of the Pivotal Phase III MDD Program

The cornerstone of the MDD program consisted of three large, multicenter, randomized, double-blind, placebo-controlled trials: LNBM (NCT01173601), LNBQ (NCT01187407), and LNBR (NCT01185340).[5]

Study Design and Population: The trials shared a sophisticated design intended to isolate a true partial-responder population. Patients first entered a 3-week, single-blind placebo lead-in phase while continuing their SSRI. Only those who demonstrated a continued partial response (defined as <25% improvement on the Montgomery-Asberg Depression Rating Scale and a total score ≥14) were then randomized to receive either Edivoxetine (at various fixed or flexible doses) or a matching placebo, in addition to their ongoing SSRI, for an 8-week double-blind treatment period.[6]

Primary Endpoint and Results: The primary efficacy endpoint across all three studies was the mean change in the MADRS total score from the randomization baseline to the end of the 8-week treatment period.[5] The results were unequivocally negative. In each of the three trials, Edivoxetine failed to demonstrate a statistically significant separation from placebo.

The data, summarized in Table 2, reveal a critical finding: an exceptionally high placebo response. In all arms, patients who received an adjunctive placebo experienced a substantial improvement in their depressive symptoms, with mean MADRS score reductions ranging from -7.8 to -9.4 points.[6] This powerful placebo effect created a therapeutic ceiling that Edivoxetine was unable to surpass. The difference in improvement between the active drug and placebo was clinically and statistically non-significant across all doses tested. This consistent failure across three large, well-designed trials provided definitive evidence against the efficacy of this treatment strategy.

Table 2: Summary of Efficacy Outcomes from the Three Pivotal Phase III Adjunctive MDD Trials

Study IdentifierTreatment ArmNLS Mean Change from Baseline in MADRS Score
Study 1 (LNBM)Edivoxetine 12 mg + SSRI231-8.5
Edivoxetine 18 mg + SSRI230-8.7
Placebo + SSRI240-7.8
Study 2 (LNBQ)Edivoxetine 6 mg + SSRI226-9.6
Edivoxetine 12-18 mg + SSRI232-9.4
Placebo + SSRI231-9.4
Study 3 (LNBR)Edivoxetine 12-18 mg + SSRI230-8.7
Placebo + SSRI219-8.5
Data sourced from a publication detailing the results of the three trials.6 LS = Least-Squares.

The design of these trials, while rigorous, may have inadvertently contributed to their failure. Selecting for partial responders and then introducing a new "add-on" treatment is a psychologically potent intervention. The act of enrollment and the expectation of benefit from a novel agent can drive a powerful placebo response, a phenomenon sometimes referred to as the "adjunctive design paradox." The trials were not merely testing Edivoxetine against an inert pill; they were testing it against the powerful therapeutic effect of adding a new treatment to an existing one, a hurdle it could not clear.

The Maintenance Phase: Failure to Prevent Symptom Re-emergence

To further assess its utility, a separate relapse-prevention study (NCT01299272) was conducted. In this trial, patients who achieved remission after an open-label phase of adjunctive Edivoxetine treatment were randomized to either continue the drug or be switched to placebo.[12] This withdrawal design is a stringent test of a drug's maintenance effect.

The results of this study were also negative. There was no significant difference in the primary outcome, the time to re-emergence of depressive symptoms, between the Edivoxetine and placebo groups (Kaplan-Meier log-rank p = 0.485).[12] The failure to prevent relapse in patients who had previously responded to the drug provided the final piece of evidence against its efficacy in MDD.

Investigation in Attention-Deficit/Hyperactivity Disorder (ADHD)

Parallel to the MDD program, Edivoxetine was investigated as a non-stimulant treatment for ADHD, a logical application for an NRI given the established role of norepinephrine in regulating attention and executive function.[1]

A large, randomized, double-blind, placebo-controlled study (NCT00922636) in 340 pediatric patients (ages 6–17) yielded initially promising results.[24] After 8 weeks of treatment, Edivoxetine at doses of 0.2 mg/kg/day and 0.3 mg/kg/day demonstrated a statistically significantly greater improvement in ADHD symptoms compared to placebo, as measured by the ADHD Rating Scale (ADHD-RS) total score (p<0.010 for both doses).[21] The calculated effect sizes of 0.51 and 0.54 were considered moderate and clinically meaningful, approaching the effect size of 0.69 observed for the active comparator, OROS methylphenidate, in a subset of patients.[21] These findings suggested that Edivoxetine could be a viable therapeutic option for ADHD.

Re-evaluation of ADHD Efficacy: Insights from Contemporary Meta-Analyses

The initial optimism surrounding Edivoxetine for ADHD has been significantly tempered by more recent, higher-level evidence. A forthcoming (August 2024 publication) Bayesian network meta-analysis, which represents a comprehensive synthesis of data from 31 relevant clinical trials of various monoamine reuptake inhibitors in ADHD, reached a starkly different conclusion.[4]

This extensive analysis found that Edivoxetine was no better than placebo in reducing the severity of ADHD symptoms.[4] This contradiction between the findings of a single pivotal trial and a large-scale meta-analysis is a critical development. Network meta-analyses are considered a higher level of evidence because they integrate both direct and indirect comparisons across a wide range of studies, providing a more robust and contextualized estimate of a drug's true effect. The negative finding from this analysis suggests that the positive result from the single trial may have been a statistical anomaly (a Type I error) or an effect that is not replicable or generalizable. This likely explains why, despite a seemingly "positive" Phase III study, the development of Edivoxetine for ADHD was also not pursued for regulatory submission.

Comprehensive Safety and Tolerability Assessment

The safety profile of an investigational drug is as crucial as its efficacy. For Edivoxetine, the adverse events observed in clinical trials were highly consistent with its known mechanism of action, providing further evidence of its biological activity while also defining its clinical risk profile.

Profile of Treatment-Emergent Adverse Events (TEAEs)

A pooled analysis of data from four placebo-controlled adjunctive MDD trials, encompassing 1254 patients treated with Edivoxetine and 801 with placebo, provides a robust overview of its short-term tolerability.[7]

Overall, a significantly higher percentage of patients in the Edivoxetine group reported at least one TEAE compared to the placebo group (56.8% vs. 43.7%, p<0.001).[7] The most frequently reported TEAEs (≥5% incidence) were hyperhidrosis (excessive sweating), nausea, and tachycardia.[7] Discontinuation due to adverse events was numerically higher in the Edivoxetine group (4.9%) than the placebo group (3.5%), though this difference was not statistically significant (p=0.07).[7]

Table 3 details the TEAEs that occurred with an incidence of at least 2% and were significantly more frequent with Edivoxetine than placebo. This pattern of events is not random but forms a clear "noradrenergic signature." Long-term open-label studies in both MDD and ADHD for up to 5 years confirmed that this safety profile remained consistent over time, with no unexpected adverse events emerging with prolonged exposure.[25]

Table 3: Incidence of Common Treatment-Emergent Adverse Events (≥2%) in Pooled Adjunctive MDD Trials (Edivoxetine vs. Placebo)

Adverse EventAdjunctive Edivoxetine (N=1254) n (%)Adjunctive Placebo (N=801) n (%)p-value
Hyperhidrosis95 (7.6)9 (1.1)<0.001
Nausea79 (6.3)20 (2.5)<0.001
Tachycardia66 (5.3)3 (0.4)<0.001
Dizziness53 (4.2)17 (2.1)0.007
Vomiting37 (3.0)7 (0.9)<0.001
Constipation35 (2.8)10 (1.3)0.020
Palpitations30 (2.4)1 (0.1)<0.001
Testicular pain (males)10 (2.3)00.010
Dry mouth28 (2.2)9 (1.1)0.046
Increased heart rate26 (2.1)1 (0.1)<0.001
Data sourced from a pooled analysis of four placebo-controlled trials.7

The Noradrenergic Signature: Cardiovascular and Autonomic Effects

The most clinically significant adverse effects of Edivoxetine were directly related to its intended pharmacological action of increasing synaptic norepinephrine.

Cardiovascular Effects: Consistent across all studies in both MDD and ADHD, treatment with Edivoxetine was associated with statistically significant mean increases in systolic blood pressure, diastolic blood pressure, and pulse rate.[2] In the pooled MDD data, the mean increase in sitting pulse was 8.8 bpm with Edivoxetine versus a decrease of 1.3 bpm with placebo (p<0.001).[7] While these changes were generally modest on average, they necessitated monitoring and represented a potential risk for patients with pre-existing cardiovascular conditions. These effects were a direct consequence of norepinephrine's action on adrenergic receptors in the cardiovascular system.

Autonomic Effects: Other classic noradrenergic side effects were also prominent. Hyperhidrosis, dry mouth, and constipation are all mediated by the sympathetic nervous system, which is activated by norepinephrine.[2] The high incidence of these TEAEs further confirms that Edivoxetine was exerting a potent, systemic noradrenergic effect. This safety profile serves as a powerful confirmation of the drug's mechanism; it was active enough to produce these expected side effects, which makes its lack of therapeutic efficacy all the more definitive.

Clinically Significant Drug-Drug Interactions

The potential for drug-drug interactions with Edivoxetine stems from both its pharmacodynamic and pharmacokinetic properties.

Pharmacodynamic Interactions: As a CNS-active agent, Edivoxetine was flagged for a potential increase in the risk and severity of CNS depression when combined with a wide array of other CNS depressants. This includes benzodiazepines (e.g., clonazepam), opioids (e.g., alfentanil), tricyclic antidepressants (e.g., amitriptyline), antipsychotics, muscle relaxants, and even substances like alcohol and cannabidiol.[1] While this is a general cautionary note for many psychotropic drugs, the breadth of the list would have presented a significant clinical management challenge, given the high rates of polypharmacy in patients with MDD.

Pharmacokinetic Interactions: The metabolism of Edivoxetine via CYP2D6 and CYP3A4 creates a high potential for pharmacokinetic interactions.[22] Co-administration with potent inhibitors of these enzymes (e.g., bupropion, fluoxetine, paroxetine for CYP2D6; ketoconazole, clarithromycin for CYP3A4) would be expected to increase Edivoxetine plasma concentrations, potentially exacerbating its cardiovascular and other side effects. Conversely, co-administration with strong inducers (e.g., carbamazepine, rifampin) could decrease its plasma levels, leading to a loss of effect.

Discontinuation of Edivoxetine: A Post-Mortem Analysis

The decision to terminate the development of a late-stage clinical candidate is a significant event for a pharmaceutical company and for the medical field it aims to serve. The discontinuation of Edivoxetine provides a clear and instructive case study on the complex interplay between pharmacology, clinical trial design, and the high bar for demonstrating efficacy in modern psychiatry.

The Official Rationale: A Clear Failure of Efficacy

On December 5, 2013, Eli Lilly and Company publicly announced the discontinuation of the clinical development of Edivoxetine for the adjunctive treatment of MDD.[5] The company's rationale was direct and unambiguous, resting entirely on the primary outcome of the Phase III program. In the official press release, David Ricks, then president of Lilly Bio-Medicines, stated, "the lack of efficacy compared to SSRI alone in three separate clinical trials means that Lilly will not proceed with development of edivoxetine as an add-on treatment for depression".[5]

The company acknowledged that the safety and tolerability profile was consistent with previous studies and its known mechanism of action. However, without a demonstrable efficacy benefit, this acceptable safety profile was insufficient to support a regulatory submission.[5] The decision was expected to result in a fourth-quarter R&D charge of approximately $15 million (pre-tax), a figure that represents only a fraction of the total investment in a multi-year, multi-trial Phase III program.[8]

Contextualizing the Failure: The Enduring Challenge of Placebo Response in CNS Trials

Eli Lilly's announcement pointedly contextualized the failure within the broader challenges of neuroscience research, specifically citing the "historically high placebo response rate" as a prevalent issue.[5] This acknowledgment is critical to understanding the outcome. The Edivoxetine MDD trials can be viewed as a classic example of "failed" trials rather than "negative" trials.[28] A truly negative trial implies that the drug is inert or ineffective. A failed trial, however, is one in which the study design and high levels of non-specific therapeutic effects (i.e., "noise") make it impossible to detect a true, albeit potentially modest, drug effect.

The placebo response rates in the Edivoxetine trials, with MADRS score improvements of 8 to 9 points, were exceptionally high.[6] This phenomenon is multifactorial and is known to be influenced by patient expectation, the intensity of clinical contact, and specific trial design features.[28] As discussed, the adjunctive design used for Edivoxetine is particularly susceptible to inflating these effects. Therefore, the central narrative of Edivoxetine's failure is not simply that the drug "did not work." A more nuanced conclusion is that the drug did not provide a therapeutic benefit powerful enough to be detected above the substantial improvement seen in patients receiving a placebo in the specific context of this rigorous clinical trial program. The drug was biologically active, as shown by its pharmacodynamic biomarker effects and its side effect profile, but this activity did not translate into a clinically superior outcome.

Implications for the Future of Selective NRI Development in Psychiatry

The definitive failure of Edivoxetine has had significant implications for the field. As a potent and highly selective agent, its inability to demonstrate efficacy casts considerable doubt on the viability of the selective NRI mechanism as a robust strategy for treating MDD, particularly in the large population of SSRI partial responders. The result lends support to the hypothesis that broader neurochemical modulation, such as that provided by dual-action SNRIs or other multi-target agents, may be required to achieve superior antidepressant efficacy.[29]

At the time, the failure of Edivoxetine, along with other setbacks, left Eli Lilly's late-stage CNS pipeline nearly empty, underscoring the high-risk, high-attrition nature of neuroscience drug development.[32] The story of Edivoxetine serves as a powerful reminder that a plausible mechanism and a well-designed molecule are no guarantee of clinical success. It highlights the urgent need for more innovative clinical trial designs that can mitigate the placebo effect and for better translational biomarkers that can more accurately predict clinical efficacy from preclinical and early-phase data.

Comparative Analysis: Edivoxetine in the Context of Other Selective NRIs

To fully appreciate the clinical and scientific standing of Edivoxetine, it is essential to place it in the context of its closest pharmacological relatives: atomoxetine and reboxetine. This comparison reveals important patterns regarding the therapeutic utility of the selective NRI mechanism across different psychiatric disorders.

A structured comparison of these three agents across key domains is presented in Table 4.

Table 4: Comparative Profile of Selective NRIs: Edivoxetine vs. Atomoxetine vs. Reboxetine

FeatureEdivoxetineAtomoxetineReboxetine
Primary Indication(s)Investigational for MDD & ADHD 1Attention-Deficit/Hyperactivity Disorder (ADHD) 33Major Depressive Disorder (MDD) 33
Regulatory StatusDevelopment Discontinued; Not Approved 2Approved in US & Europe (Strattera) 33Approved in Europe (Edronax); Not Approved in US 33
Efficacy in MDDConclusively failed to show efficacy in multiple Phase III trials 5Not a primary indication; may be used off-label with comorbid ADHD 35Highly controversial; some meta-analyses show no benefit over placebo 34
Efficacy in ADHDInitial positive trial contradicted by later meta-analysis showing no benefit 10Established efficacy as a non-stimulant treatment option 37Evidence supports efficacy; used off-label 34
Key PK (t1/2​)~4-6 hours 11~3.6 hours (extensive metabolizers) 33~12-13 hours 33
Common Noradrenergic Side EffectsIncreased BP/pulse, hyperhidrosis, nausea, dry mouth 7Dry mouth, insomnia, nausea, decreased appetite, cardiovascular effects 33Insomnia, dry mouth, constipation, tachycardia, urinary hesitancy 33
Overall Clinical StandingA definitive clinical development failureAn established, niche second-line therapy for ADHDA controversial antidepressant with limited use and questionable efficacy

This comparative analysis reveals a striking pattern of indication specificity for the selective NRI mechanism.

Atomoxetine has successfully carved out a durable clinical niche as a non-stimulant treatment for ADHD.[33] Its efficacy, while generally considered more modest than that of stimulants, is well-established, and it provides a valuable alternative for patients who cannot tolerate stimulants or for whom there is a concern for substance abuse.[33]

Reboxetine, in contrast, has had a tumultuous history. While approved for MDD in many European countries, its efficacy has been the subject of intense debate. A highly publicized 2010 meta-analysis that included unpublished data from Pfizer concluded that reboxetine was ineffective and potentially harmful.[34] While subsequent reviews have suggested it may have benefit in severe depression, its clinical utility remains contested, and it failed to gain approval in the United States.[33]

Edivoxetine represents the most definitive failure of the three in the mood disorders space. Its large, robust Phase III program provided a clear and convincing negative result for adjunctive MDD treatment.[5] Furthermore, its potential in ADHD, initially supported by a single trial, has been effectively nullified by more comprehensive meta-analytic evidence.[10]

This pattern strongly suggests that while enhancing noradrenergic tone via selective NET inhibition is a viable and effective strategy for modulating the neural circuits underlying attention and executive function in ADHD, it is an insufficient and unreliable mechanism for robustly treating the complex pathophysiology of major depression. The clinical success of atomoxetine in ADHD alongside the failures of Edivoxetine and the controversy surrounding reboxetine in MDD provides a crucial lesson in matching a specific pharmacological mechanism to the correct disease state.

Conclusion and Final Perspectives

Edivoxetine (LY-2216684) emerged from a rational drug design program as a potent, highly selective, and pharmacologically active norepinephrine reuptake inhibitor. It possessed a predictable pharmacokinetic profile and demonstrated clear evidence of biological target engagement in human subjects, as measured by its effect on the DHPG biomarker and its mechanism-consistent safety profile.

Despite this solid pharmacological foundation, the Edivoxetine development program culminated in a definitive clinical failure. The extensive and well-designed Phase III program for Major Depressive Disorder conclusively demonstrated that, as an adjunctive therapy for patients with a partial response to SSRIs, Edivoxetine offered no significant therapeutic benefit over placebo. This lack of efficacy was underscored by a failure to prevent symptom re-emergence in a maintenance study. The initially promising data for its use in ADHD has also been convincingly refuted by a subsequent, more comprehensive meta-analysis, solidifying its status as an investigational drug without a viable clinical application.

The narrative of Edivoxetine's failure is not one of an inert or unsafe compound. Rather, it is a story of a clear disconnect between biological activity and clinical efficacy. The drug was working as intended at a molecular and systemic level, but this action failed to translate into a meaningful improvement in patient outcomes that could be distinguished from the powerful non-specific effects captured by the placebo arm, particularly within the challenging context of an adjunctive therapy trial design.

The legacy of the Edivoxetine program is therefore that of a crucial, albeit expensive, scientific experiment. It provided a clear and valuable negative answer to an important clinical question: is selective noradrenergic augmentation a robust strategy for the millions of patients who only partially respond to first-line serotonergic antidepressants? The data from the Edivoxetine trials strongly suggest that it is not. Its story serves as a powerful case study in the immense challenges of CNS drug development, highlighting the persistent problem of the placebo response, the risks of trial designs that may inadvertently inflate it, and the sobering reality that even the most precisely engineered molecules can fail at the final clinical hurdle. The lessons learned from the development and discontinuation of Edivoxetine continue to inform strategies for the discovery and validation of the next generation of treatments for psychiatric disorders.

Works cited

  1. Edivoxetine: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed September 7, 2025, https://go.drugbank.com/drugs/DB09184
  2. Edivoxetine - Wikipedia, accessed September 7, 2025, https://en.wikipedia.org/wiki/Edivoxetine
  3. What is Edivoxetine Hydrochloride used for? - Patsnap Synapse, accessed September 7, 2025, https://synapse.patsnap.com/article/what-is-edivoxetine-hydrochloride-used-for
  4. Edivoxetine Hydrochloride - Drug Targets, Indications, Patents - Synapse, accessed September 7, 2025, https://synapse-patsnap-com.libproxy1.nus.edu.sg/drug/266e9498625e40659673d77fad9a8a0c
  5. Lilly Announces Edivoxetine Did Not Meet Primary Endpoint of ..., accessed September 7, 2025, https://investor.lilly.com/news-releases/news-release-details/lilly-announces-edivoxetine-did-not-meet-primary-endpoint-phase
  6. Efficacy outcomes from 3 clinical trials of edivoxetine as adjunctive treatment for patients with major depressive disorder who are partial responders to selective serotonin reuptake inhibitor treatment - PubMed, accessed September 7, 2025, https://pubmed.ncbi.nlm.nih.gov/27035159/
  7. Safety and tolerability of edivoxetine as adjunctive treatment to selective serotonin reuptake inhibitor antidepressants for patients with major depressive disorder - PMC, accessed September 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4434861/
  8. Lilly pulls plug on antidepressant edivoxetine - PharmaTimes, accessed September 7, 2025, https://pharmatimes.com/news/lilly_pulls_plug_on_antidepressant_edivoxetine_1006139/
  9. Lilly Announces Edivoxetine Did Not Meet Primary Endpoint of Phase III Clinical Studies as Add-On Therapy for Major Depressive Disorder - PR Newswire, accessed September 7, 2025, https://www.prnewswire.com/news-releases/lilly-announces-edivoxetine-did-not-meet-primary-endpoint-of-phase-iii-clinical-studies-as-add-on-therapy-for-major-depressive-disorder-234579581.html
  10. Edivoxetine Hydrochloride - Drug Targets, Indications, Patents - Patsnap Synapse, accessed September 7, 2025, https://synapse.patsnap.com/drug/266e9498625e40659673d77fad9a8a0c
  11. Pharmacokinetics and pharmacodynamics of edivoxetine (LY2216684), a norepinephrine reuptake inhibitor, in pediatric patients with attention-deficit/hyperactivity disorder - PubMed, accessed September 7, 2025, https://pubmed.ncbi.nlm.nih.gov/22849510/
  12. Edivoxetine compared to placebo as adjunctive therapy to selective serotonin reuptake inhibitors in the prevention of symptom re-emergence in major depressive disorder - PubMed, accessed September 7, 2025, https://pubmed.ncbi.nlm.nih.gov/25894953/
  13. Practical Asymmetric Synthesis of an Edivoxetine·HCl Intermediate via an Efficient Diazotization Process - ACS Publications, accessed September 7, 2025, https://pubs.acs.org/doi/abs/10.1021/acs.oprd.5b00014
  14. Edivoxetine | norepinephrine inhibitor | CAS# 1194508-25-2 | InvivoChem, accessed September 7, 2025, https://www.invivochem.com/edivoxetine.html
  15. Compound: EDIVOXETINE (CHEMBL2103857) - ChEMBL - EMBL-EBI, accessed September 7, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL2103857
  16. Edivoxetine (LY 2216684) | Norepinephrine Reuptake Inhibitor | MedChemExpress, accessed September 7, 2025, https://www.medchemexpress.com/edivoxetine.html
  17. CAS 1194508-25-2: Edivoxetine | CymitQuimica, accessed September 7, 2025, https://cymitquimica.com/cas/1194508-25-2/
  18. 3D structure for Edivoxetine (DB09184) | DrugBank Online, accessed September 7, 2025, https://go.drugbank.com/structures/small_molecule_drugs/DB09184
  19. en.wikipedia.org, accessed September 7, 2025, [https://en.wikipedia.org/wiki/Edivoxetine#:~:text=Edivoxetine%20(INN%3B%20code%20name%20LY,and%20as%20an%20antidepressant%20treatment.](https://www.google.com/url?q=https://en.wikipedia.org/wiki/Edivoxetine%23:~:text%3DEdivoxetine%2520(INN%253B%2520code%2520name%2520LY,and%2520as%2520an%2520antidepressant%2520treatment.&sa=D&source=editors&ust=1757241470940203&usg=AOvVaw3qkJqZCq9z6_KFxi7soTaV)
  20. Implications for Combination Therapy of Selective Monoamine Reuptake Inhibitors on Dopamine Transporters - MDPI, accessed September 7, 2025, https://www.mdpi.com/2227-9059/11/10/2846
  21. Pharmacokinetics and Pharmacodynamics of Edivoxetine ..., accessed September 7, 2025, https://www.researchgate.net/publication/230591070_Pharmacokinetics_and_Pharmacodynamics_of_Edivoxetine_LY2216684_a_Norepinephrine_Reuptake_Inhibitor_in_Pediatric_Patients_with_Attention-Deficit_Hyperactivity_Disorder
  22. A pharmacokinetic evaluation of oral edivoxetine hydrochloride for the treatment of attention deficit-hyperactivity disorder, accessed September 7, 2025, https://www.tandfonline.com/doi/pdf/10.1517/17425255.2014.939955
  23. The effect of hepatic or renal impairment on the pharmacokinetics of ..., accessed September 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3832778/
  24. A Randomized Trial of Edivoxetine in Pediatric Patients with Attention-Deficit/Hyperactivity Disorder - PMC - PubMed Central, accessed September 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4026219/
  25. (PDF) Long-Term, Open-Label, Safety Study of Edivoxetine as Adjunctive Treatment for Adult Patients with Major Depressive Disorder Who Were Partial Responders to Selective Serotonin Reuptake Inhibitor Treatment in Japan - ResearchGate, accessed September 7, 2025, https://www.researchgate.net/publication/275252256_Long-Term_Open-Label_Safety_Study_of_Edivoxetine_as_Adjunctive_Treatment_for_Adult_Patients_with_Major_Depressive_Disorder_Who_Were_Partial_Responders_to_Selective_Serotonin_Reuptake_Inhibitor_Treatme
  26. Long-Term, Open-Label, Safety Study of Edivoxetine Monotherapy in Children and Adolescents with Attention-Deficit/Hyperactivity Disorder - PubMed, accessed September 7, 2025, https://pubmed.ncbi.nlm.nih.gov/28402139/
  27. Safety and Tolerability of Edivoxetine for Long-Term Treatment of Major Depressive Disorder in Adult Patients - Scientific Research Publishing, accessed September 7, 2025, https://www.scirp.org/journal/paperinformation?paperid=45046
  28. The Impact of Failed Antidepressant Trials on Outcomes in Children ..., accessed September 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9208430/
  29. Failure of first SSRI for depression - What is the next step? - ResearchGate, accessed September 7, 2025, https://www.researchgate.net/publication/5600818_Failure_of_first_SSRI_for_depression_-_What_is_the_next_step
  30. Remission, dropouts, and adverse drug reaction rates in major ..., accessed September 7, 2025, https://www.ncbi.nlm.nih.gov/books/NBK73259/
  31. A critical review of pharmacotherapy for major depressive disorder - Oxford Academic, accessed September 7, 2025, https://academic.oup.com/ijnp/article/14/10/1417/753918
  32. Lilly's Late-Stage CNS Pipeline Bare Following Edivoxetine Failure, accessed September 7, 2025, https://insights.citeline.com/PS075224/Lillys-Late-Stage-CNS-Pipeline-Bare-Following-Edivoxetine-Failure/
  33. Selective norepinephrine reuptake inhibitor - Wikipedia, accessed September 7, 2025, https://en.wikipedia.org/wiki/Selective_norepinephrine_reuptake_inhibitor
  34. Reboxetine - Wikipedia, accessed September 7, 2025, https://en.wikipedia.org/wiki/Reboxetine
  35. Comparison between Atomoxetine and OROS Methylphenidate as an Adjunctive to SSRIs in Attention-deficit/Hyperactivity Disorder Adults with Comorbid Partially Responsive Major Depressive Disorder: A Head-to-head, 12-week, Randomized, Rater-blinded Clinical Trial, accessed September 7, 2025, https://www.cpn.or.kr/journal/view.html?doi=10.9758/cpn.2022.20.1.143
  36. Norepinephrine reuptake inhibitor - Wikipedia, accessed September 7, 2025, https://en.wikipedia.org/wiki/Norepinephrine_reuptake_inhibitor
  37. Meta-analysis on the efficacy of the norepinephrine reuptake inhibitors reboxetine and atomoxetine for the treatment of schizophrenia and attention deficit hyperactivity disorder - Advances in Clinical and Experimental Medicine, accessed September 7, 2025, https://advances.umw.edu.pl/pdf/2023/32/5/511.pdf
  38. Efficacy of reboxetine in adults with attention-deficit/hyperactivity disorder: A randomized, placebo-controlled clinical trial | Request PDF - ResearchGate, accessed September 7, 2025, https://www.researchgate.net/publication/49827776_Efficacy_of_reboxetine_in_adults_with_attention-deficithyperactivity_disorder_A_randomized_placebo-controlled_clinical_trial

Published at: September 7, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.