C17H21NO
83015-26-3
Attention Deficit Hyperactivity Disorder (ADHD)
Atomoxetine is a small molecule drug that occupies a unique and significant position in the therapeutic armamentarium for Attention-Deficit/Hyperactivity Disorder (ADHD).[1] It was the first non-stimulant medication to receive approval from the U.S. Food and Drug Administration (FDA) for this indication, representing a paradigm shift in treatment options for a condition historically managed with psychostimulants.[1] Classified pharmacologically as a selective norepinephrine reuptake inhibitor (SNRI), its mechanism is distinct from that of methylphenidate and amphetamine-based medications.[2]
Atomoxetine, identified by DrugBank Accession Number DB00289 and CAS Number 83015-26-3, is indicated for the management of ADHD in children aged six years and older, adolescents, and adults.[1] Its use is intended as a component of a comprehensive treatment program that integrates psychological, educational, and social interventions to address the multifaceted nature of the disorder.[1] The development of atomoxetine was driven by the clinical need for an effective ADHD therapy devoid of the abuse potential and diversion risks associated with traditional stimulant medications. Consequently, its primary clinical utility is often realized in patients for whom stimulants are contraindicated, ineffective, or poorly tolerated, or in cases where there is a significant concern for substance misuse.[4]
Atomoxetine is a phenoxy-3-propylamine derivative, specifically a secondary amino compound with the IUPAC name (3R)-N-methyl-3-(2-methylphenoxy)-3-phenylpropan-1-amine.[5] The therapeutic activity resides in the (R)-(-) stereoisomer, which is approximately nine times more potent as a norepinephrine reuptake inhibitor than its S(+) counterpart.[3] In clinical and commercial settings, atomoxetine is almost exclusively formulated and administered as its hydrochloride salt, atomoxetine hydrochloride (CAS: 82248-59-7), which appears as a white to off-white crystalline powder.[1] The parent compound is soluble in organic solvents such as dimethyl sulfoxide (DMSO), ethanol, and dimethylformamide (DMF).[13]
The development and regulatory approval of atomoxetine can be seen as a direct strategic response to the primary limitations of the dominant ADHD drug class. At a time of growing concern over the abuse and diversion of stimulant medications, a non-controlled substance with a novel mechanism offered a significant clinical and commercial advantage. Its entire therapeutic profile—from its slower onset of action to its lack of euphoriant effects—is defined by its neurochemical distinction from stimulants. This distinction underpins its value as an alternative therapy and frames the risk-benefit analysis for clinicians and patients.
Table 1: Drug Identification and Chemical Properties of Atomoxetine
Property | Value | Source(s) |
---|---|---|
Common Name | Atomoxetine | 1 |
IUPAC Name | (3R)-N-methyl-3-(2-methylphenoxy)-3-phenylpropan-1-amine | 5 |
DrugBank ID | DB00289 | 1 |
CAS Number (Parent) | 83015-26-3 | 5 |
CAS Number (HCl Salt) | 82248-59-7 | 4 |
Molecular Formula | C17H21NO | 5 |
Molecular Weight (Parent) | 255.35 g/mol | 14 |
Molecular Weight (HCl Salt) | 291.82 g/mol | 5 |
InChI | InChI=1S/C17H21NO/c1-14-8-6-7-11-16(14)19-17(12-13-18-2)15-9-4-3-5-10-15/h3-11,17-18H,12-13H2,1-2H3/t17-/m1/s1 | 5 |
InChIKey | VHGCDTVCOLNTBX-QGZVFWFLSA-N | 5 |
Canonical SMILES | CNCC[C@H](C1=CC=CC=C1)OC2=CC=CC=C2C | 14 |
Drug Class | Norepinephrine Reuptake Inhibitor; Adrenergic Uptake Inhibitor | 1 |
ATC Code | N06BA09 | 4 |
Atomoxetine was developed, manufactured, and originally marketed by Eli Lilly and Company under the brand name Strattera.[1] It received its initial FDA approval for the treatment of ADHD on November 26, 2002.[4] This approval marked a significant milestone, introducing the first major non-stimulant pharmacological agent for ADHD into the U.S. market.
Eli Lilly's patent for Strattera faced legal challenges, but the company ultimately prevailed in litigation, delaying the entry of generic competitors.[4] The patent expired in May 2017, and the FDA subsequently approved generic versions of atomoxetine from several pharmaceutical manufacturers, increasing market competition and accessibility.[4] A key regulatory feature of atomoxetine is its classification as a non-controlled substance by the Drug Enforcement Administration (DEA).[5] This status, which contrasts sharply with the Schedule II classification of all stimulant ADHD medications, reflects its low potential for abuse and dependence and simplifies the prescribing process for healthcare providers.[18]
The therapeutic effects of atomoxetine are primarily attributed to its function as a potent and highly selective inhibitor of the presynaptic norepinephrine transporter (NET), which is encoded by the SLC6A2 gene.[2] By binding to NET with high affinity (
Ki = 5 nM), atomoxetine blocks the reuptake of norepinephrine (NE) from the synaptic cleft into the presynaptic neuron.[14] This action increases the concentration and prolongs the availability of NE in the extracellular space, enhancing noradrenergic neurotransmission.[23]
While the official labeling states that the precise mechanism of action is unknown, the downstream neurochemical consequences of NET inhibition are well-characterized.[1] In animal models, atomoxetine administration leads to a roughly three-fold increase in extracellular NE levels in the prefrontal cortex (PFC).[14] Critically, this NET inhibition in the PFC also results in a secondary, localized increase in extracellular dopamine (DA) levels.[2] This occurs because in the PFC, which has a low density of dopamine transporters (DAT), DA reuptake is primarily mediated by NET. By blocking NET, atomoxetine effectively increases the availability of both key catecholamines—NE and DA—in this cortical region crucial for executive functions like attention, impulse control, and working memory.
This regional selectivity is the neurobiological foundation of atomoxetine's favorable safety profile concerning abuse liability. Unlike psychostimulants, atomoxetine does not significantly increase dopamine in subcortical reward centers such as the nucleus accumbens and striatum.[7] These regions are densely populated with DAT, and their dopaminergic activation is strongly associated with the reinforcing and addictive properties of drugs of abuse.[2] Atomoxetine's minimal activity at DAT (
Ki = 1,451 nM) prevents this widespread dopaminergic surge, thereby dissociating its therapeutic effects from abuse potential.[14] Its selectivity for NET is substantially greater than for the serotonin transporter (SERT,
Ki = 77 nM) and other neurotransmitter receptors, distinguishing it from less selective agents like tricyclic antidepressants.[1]
More recent investigations suggest a broader pharmacodynamic profile. Atomoxetine has been shown to act as an antagonist at the N-methyl-D-aspartate (NMDA) receptor, implicating a potential modulatory role in the glutamatergic system.[7] Furthermore, it directly inhibits hERG potassium ion channels (
IC50 = 6.3 µM), an action that underlies the risk of cardiac QT interval prolongation and potential arrhythmias.[4]
Following oral administration, atomoxetine is rapidly absorbed, with peak plasma concentrations (Tmax) reached in 1-2 hours.[1] The presence of food does not significantly affect the overall extent of absorption (AUC) but can delay Tmax by approximately 3 hours and reduce the peak concentration (Cmax).[1] Atomoxetine is extensively distributed and highly bound (approximately 98%) to plasma proteins, predominantly albumin.[1]
The metabolism of atomoxetine is the most critical aspect of its pharmacokinetics and is dominated by the activity of the cytochrome P450 2D6 (CYP2D6) enzyme.[1] This enzyme catalyzes the primary metabolic pathway, hydroxylation, to form the major active metabolite, 4-hydroxyatomoxetine.[1] This metabolite is equipotent to the parent drug as a NET inhibitor but typically circulates at much lower concentrations before being rapidly inactivated through glucuronidation.[1] Minor metabolic pathways, such as N-demethylation via other CYP enzymes (e.g., CYP2C18), contribute minimally to the overall clinical effect, particularly in individuals with normal CYP2D6 function.[22]
The polymorphic nature of the CYP2D6 gene leads to profound inter-individual variability in atomoxetine metabolism, effectively creating two distinct pharmacokinetic profiles within the patient population. The distinction between CYP2D6 "Extensive Metabolizers" (EMs), who have normal enzyme function, and "Poor Metabolizers" (PMs), who have deficient enzyme function, is the single most important variable governing the drug's in vivo behavior. A standard dose for an EM patient can result in a significant overdose for a PM patient. This bimodal distribution of drug exposure means that the "average" patient response reported in clinical trials is a statistical composite of two very different groups. This reality underscores the fundamental link between an individual's genetic makeup and the drug's safety and efficacy, highlighting the potential clinical utility of pharmacogenomic testing to personalize atomoxetine therapy and mitigate risks.
In EMs, who constitute the majority of the population, atomoxetine is cleared efficiently. In contrast, PMs (approximately 7% of Caucasians) experience dramatically higher drug exposure. As detailed in Table 2, PMs exhibit a 10-fold greater area under the curve (AUC) and a 5-fold greater Cmax compared to EMs.[27] This disparity is driven by a significantly prolonged elimination half-life, which is approximately 5 hours in EMs but extends to over 20 hours in PMs.[22]
Over 80% of an administered dose is excreted in the urine, almost entirely as metabolites, with the 4-hydroxyatomoxetine-O-glucuronide conjugate being the most abundant. Less than 3% of the dose is excreted as unchanged parent drug, underscoring the completeness of its biotransformation.[27]
Table 2: Key Pharmacokinetic Parameters of Atomoxetine in CYP2D6 Extensive (EM) and Poor (PM) Metabolizers
Parameter | Extensive Metabolizers (EMs) | Poor Metabolizers (PMs) | Source(s) |
---|---|---|---|
Bioavailability (Absolute) | ~63% | ~94% | 1 |
Elimination Half-life (t1/2) | ~5.2 hours | ~21.6 hours | 22 |
Apparent Plasma Clearance | ~0.35 L/hr/kg | ~0.03 L/hr/kg | 27 |
Area Under Curve (AUC) | Baseline (1x) | ~10-fold higher | 27 |
Peak Concentration (Cmax) | Baseline (1x) | ~5-fold higher | 27 |
Metabolite Profile (4-hydroxyatomoxetine) | 1% of parent drug concentration | 0.1% of parent drug concentration | 1 |
Atomoxetine is indicated by the FDA for the treatment of ADHD in children (age 6 and older), adolescents, and adults.[3] Its efficacy has been rigorously established in a series of randomized, double-blind, placebo-controlled clinical trials.
In the pediatric population, efficacy was demonstrated in four acute trials lasting 6 to 9 weeks and one long-term maintenance trial.[29] The primary outcome in these studies was the change from baseline in the total score of the ADHD Rating Scale-IV-Parent Version (ADHDRS-IV-Parent:Inv). These trials consistently showed that atomoxetine, at doses of approximately 1.2 mg/kg/day, produced a statistically significant improvement in ADHD symptoms compared to placebo.[29] A dose of 1.8 mg/kg/day did not confer additional benefit over the 1.2 mg/kg/day dose. Efficacy was established for both once-daily and twice-daily dosing regimens, providing flexibility in administration.[29] The long-term maintenance study further demonstrated that patients who continued on atomoxetine after an initial response had a significantly longer time to relapse compared to those who were switched to placebo, supporting its utility for extended treatment periods.[29]
In the adult population, efficacy was established in two identical 10-week, placebo-controlled studies.[29] The primary endpoint was the change in the 18-item Total ADHD Symptom score from the Conners' Adult ADHD Rating Scale (CAARS). In both trials, adults treated with atomoxetine (titrated to a mean final dose of approximately 95 mg/day) showed statistically significant improvements in their ADHD symptoms compared to those receiving placebo.[29]
Systematic reviews and meta-analyses have generally supported these findings, with some concluding that atomoxetine's efficacy is comparable to that of immediate-release methylphenidate.[4] However, other analyses suggest that long-acting stimulant formulations, such as OROS methylphenidate, may have a greater effect size.[10]
A defining clinical characteristic of atomoxetine is its delayed onset of action, which contrasts sharply with the rapid effects of psychostimulants.[17] While stimulants typically produce noticeable effects within an hour of administration, atomoxetine requires a period of consistent daily dosing to achieve its full therapeutic potential.[30] Patients and clinicians may observe initial improvements in symptoms within the first one to two weeks of treatment.[8] However, significant clinical improvement often takes four to six weeks to become apparent, with the maximal therapeutic effect potentially not being reached for up to 8 to 12 weeks.[23] This gradual onset is a critical point for patient education to manage expectations and ensure treatment adherence. Once therapeutic levels are achieved, atomoxetine provides continuous, 24-hour symptom control, avoiding the end-of-dose "rebound" effects that can be associated with shorter-acting stimulants.[23]
Beyond its primary indication for ADHD, atomoxetine has been explored for other conditions, though these uses are not FDA-approved. It is sometimes prescribed off-label for the management of treatment-resistant depression in adults, often as an adjunctive therapy.[3] There is also literature supporting its use for cognitive disengagement syndrome (CDS) and for treating cognitive and frontal lobe symptoms associated with traumatic brain injury (TBI), such as attentional problems and fatigue.[4]
Ongoing research continues to investigate the broader potential of atomoxetine. Active clinical trials are exploring its neurobiological effects through multimodal imaging (NCT05229627), its impact on diurnal cortisol profiles (NCT03075579), and its comparative effects versus stimulants on reward processing in youth at high risk for substance use disorders (NCT03781765), a population for whom a non-stimulant may be particularly advantageous.[33]
The safety profile of atomoxetine is well-defined and is fundamentally linked to its primary mechanism of enhancing noradrenergic tone. The systemic consequences of this neurochemical action explain the majority of its adverse effects, warnings, and contraindications. Managing risk with atomoxetine is therefore not about avoiding a list of unrelated side effects, but about anticipating and managing the predictable systemic impact of noradrenergic potentiation. This unified understanding clarifies why screening for cardiovascular disease and bipolar disorder is not arbitrary but is essential to its safe use.
The tolerability of atomoxetine is a key consideration in clinical practice. The most frequently reported adverse reactions differ slightly between pediatric and adult populations, though there is considerable overlap.
Many of these side effects, particularly gastrointestinal issues and fatigue, tend to be most prominent early in treatment and may diminish over time with continued use.[35]
Table 4: Common Adverse Reactions Reported in Clinical Trials (>5% and >Placebo)
Adverse Reaction | Pediatric Patients (Atomoxetine %) | Pediatric Patients (Placebo %) | Adult Patients (Atomoxetine %) | Adult Patients (Placebo %) | Source(s) |
---|---|---|---|---|---|
Decreased Appetite | 16% | 4% | 23% | 6% | 29 |
Nausea | 10% | 6% | 26% | 6% | 29 |
Dry Mouth | N/A | N/A | 20% | 5% | 29 |
Abdominal Pain | 18% | 13% | 7% | 4% | 29 |
Vomiting | 11% | 6% | N/A | N/A | 29 |
Fatigue | 9% | 5% | 10% | 6% | 29 |
Insomnia | N/A | N/A | 15% | 8% | 29 |
Somnolence | 8% | 4% | N/A | N/A | 29 |
Erectile Dysfunction | N/A | N/A | 8% | 1% | 29 |
Urinary Hesitation/Retention | N/A | N/A | 6% | 1% | 29 |
Atomoxetine carries a United States FDA boxed warning regarding an increased risk of suicidal ideation in children and adolescents.[2] An analysis of pooled data from short-term pediatric clinical trials revealed that approximately 0.4% of atomoxetine-treated patients reported suicidal thoughts, compared with 0% of placebo-treated patients.[18] This finding necessitates vigilant monitoring of all pediatric patients for clinical worsening, the emergence of suicidal thoughts or behaviors, and any unusual changes in behavior. This monitoring should be most intensive during the initial few months of therapy and following any changes in dose, whether an increase or decrease.[2]
Clinically significant drug-drug interactions with atomoxetine are primarily driven by its metabolism via CYP2D6 and its pharmacodynamic effects on the noradrenergic system.
Table 5: Clinically Significant Drug Interactions with Atomoxetine
Interacting Drug/Class | Mechanism of Interaction | Clinical Consequence | Management Recommendation | Source(s) |
---|---|---|---|---|
Monoamine Oxidase Inhibitors (MAOIs) (e.g., phenelzine, tranylcypromine) | Pharmacodynamic Synergism | Potentially fatal reaction, including hypertensive crisis and neuroleptic malignant syndrome-like symptoms. | Contraindicated. Do not use within 14 days of each other. | 1 |
Strong CYP2D6 Inhibitors (e.g., paroxetine, fluoxetine, bupropion) | Pharmacokinetic Inhibition | Substantial increase in atomoxetine plasma exposure (AUC and Cmax) to levels seen in Poor Metabolizers, increasing risk of adverse effects. | Dose Adjustment Required. Initiate at standard starting dose but only increase to target dose after 4 weeks if well tolerated and clinically necessary. | 1 |
QTc-Prolonging Agents (e.g., thioridazine, antiarrhythmics, some antipsychotics) | Additive Pharmacodynamic Effect | Increased risk of serious cardiac arrhythmias (Torsades de Pointes). | Contraindicated or Avoid/Use with Extreme Caution. Co-administration with thioridazine is contraindicated. Caution is advised with all other QTc-prolonging drugs. | 4 |
Pressor Agents (e.g., albuterol, pseudoephedrine) | Pharmacodynamic Potentiation | Enhanced effects on blood pressure and heart rate. | Use with Caution. Monitor cardiovascular parameters closely. | 27 |
CNS Depressants (e.g., alcohol, benzodiazepines) | Additive Pharmacodynamic Effect | Potential for increased CNS depression (e.g., somnolence, dizziness). | Monitor. Advise patients of potential for additive effects. | 1 |
The use of atomoxetine is strictly contraindicated in the following situations [29]:
The dosing of atomoxetine must be individualized based on patient weight and clinical response. The FDA-approved guidelines are as follows [3]:
Atomoxetine offers flexibility in its administration [3]:
Dose adjustments are required for specific patient populations to ensure safety and efficacy.
The decision to use atomoxetine is almost always made in the context of psychostimulants, the first-line standard of care for ADHD. The comparison between these two classes reveals a series of critical trade-offs. Stimulants are generally more effective and work much faster, but this comes at the cost of abuse potential and a different side effect profile. Atomoxetine offers a safer alternative from a substance use perspective but requires patience due to its slow onset and may provide a less robust response for some individuals.
Table 6: Comparative Profile: Atomoxetine vs. Psychostimulants for ADHD
Feature | Atomoxetine | Psychostimulants (Methylphenidate/Amphetamine) | Source(s) |
---|---|---|---|
Drug Class | Selective Norepinephrine Reuptake Inhibitor (SNRI) | CNS Stimulant | 4 |
Mechanism of Action | Selective NET inhibition; increases NE and PFC DA. | DAT/NET inhibition and/or promotion of DA/NE release. | 23 |
Abuse Potential (DEA Schedule) | Non-controlled substance; very low abuse potential. | Schedule II; high potential for abuse, dependence, and diversion. | 17 |
Onset of Action | Slow: 4-8 weeks for full effect. | Rapid: <1 hour for immediate-release formulations. | 23 |
Efficacy (Effect Size) | Moderate (SMD ≈ 0.7) | High (SMD ≈ 1.0) | 9 |
Dosing Frequency | Once or twice daily; provides 24-hour coverage. | Once (ER) to multiple times (IR) daily; may have "rebound" effects. | 23 |
Withdrawal Syndrome | No | Yes (fatigue, depression, mood changes). | 17 |
Boxed Warnings | Increased risk of suicidal ideation in children/adolescents. | High potential for abuse; risk of sudden death and serious cardiovascular events. | 19 |
Place in Therapy | 2nd-line for most; 1st-line for patients with substance use history, tic disorders, or severe anxiety. | 1st-line for most patients with ADHD. | 10 |
Within the non-stimulant category, atomoxetine is often compared to the alpha-2 adrenergic agonists (guanfacine, clonidine) and the newer SNRI, viloxazine.[38] Atomoxetine and viloxazine share a similar mechanism (SNRI) and delayed onset of action. They are primarily used to target core inattentive symptoms. In contrast, the alpha-2 agonists modulate noradrenergic signaling at the postsynaptic receptor level and are often particularly effective for symptoms of hyperactivity, impulsivity, and aggression. They are frequently used as adjunctive therapy with stimulants to manage residual symptoms or side effects like insomnia.[20]
Atomoxetine is a well-established and valuable medication in the comprehensive management of ADHD. While psychostimulants remain the first-line treatment for most patients due to their rapid onset and high efficacy, atomoxetine has carved out an indispensable clinical niche.
It serves as a critical second-line agent for the significant portion of patients (20-30%) who do not respond to, or cannot tolerate, stimulant medications.[20]
More importantly, atomoxetine is a rational first-line choice in several specific clinical scenarios where the risk profile of stimulants is prohibitive. These include:
Ultimately, the selection of atomoxetine requires a nuanced, individualized clinical judgment that carefully weighs the trade-offs between efficacy, speed of action, and the specific safety and tolerability profile of the patient. Its existence provides a vital alternative that allows for the successful treatment of a broader and more complex population of individuals with ADHD.
Published at: July 21, 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.