Skip to main content
Psychopharmacology Bulletin logoLink to Psychopharmacology Bulletin
. 2024 Aug 19;54(4):45–80.

Serotonergic Drugs for the Treatment of Attention-Deficit/Hyperactivity Disorder: A Review of Past Development, Pitfalls and Failures, and a Look to the Future

Craig Chepke 1, Elizabeth Brunner 2, Andrew J Cutler 3
PMCID: PMC11385260  PMID: 39263202

Abstract

Serotonin has been implicated in the neurobiology of attention-deficit/hyperactivity disorder (ADHD) due to its association with impulsivity, attention, and emotional regulation. Many compounds with serotonergic properties have been evaluated in ADHD, but few have been approved by regulatory authorities. Utilizing a search of public databases, we identified interventions studied in ADHD. Prescribing information and peer-reviewed and gray literature helped us to determine which compounds had an underlying mechanism of action associated with changing serotonin levels. Of the 24 compounds that met the search criteria, 16 had either failed clinical studies in an ADHD population or had been discontinued from future development. The available evidence was assessed to identify the developmental history of drugs with serotonergic activity and the outlook for new ADHD drug candidates targeting serotonin. Several treatment candidates floundered due to an inability to balance effectiveness with safety, underscoring the potential importance of potency, and selectivity. Ongoing drug development includes compounds with multimodal mechanisms of action targeting neurotransmission across serotonin, norepinephrine, and dopamine pathways; it appears likely that treatment which balances competing and complementary monoamine effects may provide improved outcomes for patients. It is hoped that continuing research into ADHD treatment will produce new therapeutic options targeting the serotonergic system, which can positively impact a wide range of symptoms, including mood, anxiety, and sleep as well as attention and hyperactivity.

Keywords: adverse effects, attention deficit disorder with hyperactivity, drug development, pharmacology, serotonin, serotonin agents

Introduction

Serotonin (5-hydroxytryptamine [5-HT]) is a key molecule in human physiology, acting as a neurotransmitter within the brain and as a peripheral hormone.1 Via binding to its receptors in the central nervous system, serotonin modulates a multitude of functions, including various behaviors, mood, sleep, and cognition/memory; an imbalance or deficit within the serotonergic system can result in a range of neurobehavioral disorders, including depression, anxiety, schizophrenia, and addiction.2,3 Due to its association with attention and behaviors such as impulsivity, aggression, and behavioral inhibition,4 serotonin and the 5-HT receptor system are thought to be involved in the neurobiology of attention-deficit/hyperactivity disorder (ADHD).5 Moreover, the well-recognized link between deficits in serotonin–receptor binding and negative effects in mood disorders3,6 is consistent with the symptoms of emotional dysregulation observed in patients with ADHD,7,8 suggesting significant pathophysiological and symptomatic overlap.

ADHD is a chronic disorder characterized by symptoms of inattention and/or hyperactivity and impulsivity; these symptoms can cause significant distress to patients, as well as impaired function and quality of life.9 Data from the Global Burden of Disease study estimated an age-standardized prevalence of 1.13% in 2019, with higher rates in males (1.61%) than in females (0.63%).10 Although historically considered to be most common in children, with reported prevalence rates of up to 8.0%,11,12 recent data have suggested a similarly high prevalence among adults (up to 6.76%).11,13 There has been a large increase in the recognition, diagnosis, and burden of ADHD in the United States over the past 3 decades,14 with 10.47% of children diagnosed with ADHD in 2022, up from 6.1% in 1997.15 However, there is controversy as to whether this is due to increased symptom awareness (particularly among females),16,17 changes to the diagnostic criteria,16 recognition of higher persistence rates (>90%) into adulthood than previously supposed,18 or overmedicalization of children.19 What is clear is that individuals who are diagnosed with ADHD are at elevated risk of social difficulties,20,21 impaired academic and occupational function,22 psychiatric comorbidities,23 substance abuse,24 accidental injuries,25 and premature mortality.26

The etiology of ADHD is diverse, encompassing both environmental factors during the period of central nervous system development27 and significant genetic influences (the estimated heritability rate is approximately 80%)28; these result in alterations in brain structure and function, which impact attention, behavior, and impulse control.27 Although the exact neurobiology of ADHD remains uncertain, the neurotransmitters dopamine and norepinephrine are thought to be the key players29; dopamine is known to be involved in impulsive decision-making, motivation/reward, and risk-taking behavior,30,31 and norepinephrine with attention and arousal.32 This hypothesis was reinforced when drugs targeting the dopamine and norepinephrine pathways proved to be effective in treating ADHD.29,33 However, approximately one-quarter to one-third of patients do not respond to such therapies,3436 and the side effects associated with these treatments may restrict their use.37

Neuroanatomical evidence is consistent with a role for the serotonergic system in ADHD,38,39 while pharmacological and genetic manipulation of rodent models has implicated the 5-HT system in the hyperactive, impulsive, and mood dimensions of ADHD.40 There is an abundance of evidence from both humans and animal models relating to the involvement of serotonin receptors in the occurrence of ADHD,5,41,42 and a serotonergic hypothesis underpinning ADHD is consistent with the known association between reduced serotonin neurotransmission and mood disorders.43

Based on the assumption that direct serotonergic regulation of relevant brain development and activity influence the neurobiology of ADHD (including both core and associated symptoms), various compounds with serotonergic properties have been studied in populations with ADHD. Thus, a large selection of drugs from classes including selective serotonin reuptake inhibitors (SSRIs), serotonin and norepinephrine reuptake inhibitors (SNRIs), and tricyclic antidepressants (TCAs)—all of which target the 5-HT system—have been evaluated in clinical studies of ADHD, with varying levels of efficacy and/or safety.4448 Notably, despite a large number of clinical trials, few such compounds have been approved by regulatory agencies for the treatment of ADHD, possibly because they have inadequate activity on dopamine and/or norepinephrine.

The objective of this evidence-based narrative review is to examine the published literature to identify the developmental history of drugs with serotonergic activity that have been studied in ADHD. We aim to highlight the mechanisms of action, the highest development status—with a focus on the reasons for termination—and the possible underlying causes of why some compounds with serotonergic activity failed in this indication. We will also look at clinical studies reported to be ongoing in the first half of 2024 and assess the potential for future serotonergic treatments in ADHD.

Methods

Information Search and Data Collation

We performed a search using the ClinicalTrials.gov website to identify clinical trials in the field of ADHD. The search was conducted on 23 October 2023, using the search terms ‘ADD’ and ‘ADHD’. The resulting list of 1,610 trials was narrowed further by filtering with the term ‘interventions’. A total of 1,139 intervention trials were retrieved; these were evaluated to identify the individual drug interventions. In addition, a literature search was performed using the PubMed database, Google Scholar, and Google Patents to identify any additional drug interventions not previously captured. All drug interventions identified are listed in Supplementary Table 1.

SUPPLEMENTARY TABLE 1. Initial Interventions Identified for the Treatment of ADHD.

Generic Name(s) Brand Name(s)
2-pyridylacetic acid
Agomelatine Valdoxan, Thymanax
Amantadine Gocovri, Osmolex ER, Symmetrel
Amphetamine, dextroamphetamine sulfate, dextroamphetamine-amphetamine Adderall, Adderall XR, Mydayis, Adzenys XR-ODT, Evekeo, Dezedrine, Zenzedi, Xelstrym
Ampreloxetine, TD-9855
AR08
Atomoxetine Strattera
AZD1446
AZD3480
Bavisant, JNJ-31001074
Betahistine HCL Serc
BLI-1008
BP22042013
Bradanicline, TC-5619
Brexpiprazole Rexulti
Bupropion Wellbutrin XL, Wellbutrin SR, Aplenzin, Forfivo XL, Zyban
Cariprazine Vraylar, Reagila
Centanafadine
Clonidine HCl Clonicel
CX717
Dasotraline, SEP-225289
Desipramine Norpramin
Dexmethylphenidate Focalin
Dimethoxybenzylidene anabaseine, GTS-21, DMXBA
Divalproex Depakote
Duloxetine Cymbalta
Edivoxetine, LY2216684 n/a
Eszopiclone Lunesta
Fasoracetam, AEVI-001, LAM 105, MDGN-001, NFC 1, NS 105
Galantamine Razadyne, Reminyl
GlyTI-M
Guanfacine HCl, Guanfacine ER, SPD503, TAK-503 Tenex
Imipramine Tofranil
Lipirinen Vayarin
Lisdexamfetamine, SPD489, NRP104 Vyvanse
L-methylfolate, 5-MTHF
Lobeline
L-threonic acid magnesium salt (L-TAMS), MMFS302, MMFS202
Mazindol CR Sanorex, Mazanor
Memantine Namenda
Metadoxine Abrixone
Methamphetamine HCl Desoxyn
Methylphenidate Ritalin, Aptensio, Concerta, Contempla (NT0102), Daytrana, Jornay PM, Metadate, Quillichew, Quillivant, Rilatine, Medikinet, Adaphen, Addwize, Inspiral, Methmild, Artige, Attenta, Cognil, Equasym, Foquest, Methylin, Penid, Phenida, Prohiper, Tradea, Methylin, Adhansia
MK-0249
MK-0929
MK-8777, Org 26576, SCH 900777
Modafinil Provigil
Molindone, molindone XR, SPN-810, SPN-810M, AFX-2201, EN-1733A Moban, Zalvari
Nabiximols Sativex
Naltrexone Revia, Depade, Trexan
ND0801
Nortriptyline HCl Pamelor
NS2359, GSK372475
Oxytocin nasal spray
PDC-1421
PF-03654746
Pozanicline, ABT-089
Reboxetine Edronax
SEP-228432
Serdexmethylphenidate and dexmethylphenidate Azstarys
Sofinicline, ABT-894
Solriamfetol
SPD473/BTS 74398
TAK-137
Tipepidine, tipepidine hibenzate (in Japan) Asverin, Antupex, Asvelik, Asvex, Bitiodin, Cofdenin A, Hustel, Nodal, Sotal
Tolcapone Tasmar
Vafidemstat, ORY-2001
Varenicline Chantix
Venlafaxine Effexor, Vensir, Vencarm, Venlalix, Venlablue
Viloxazine ER, SPN-812 Qelbree
Vortioxetine Trintellix, Brintellix
Zolpidem Edluar, Ambien CR, Ambien, Intermezzo, Zolpimist

ADHD, attention-deficit/hyperactivity disorder.

Prescribing information as well as peer-reviewed and gray literature were then searched to determine which of these compounds were reported to have an underlying mechanism of action associated with the neurotransmitter serotonin or its receptor and/or transporter or altered serotonin levels via other mechanisms. We determined that 24 compounds met the criteria of being studied in ADHD and having serotonergic activity, of which 16 were found to have either failed clinical studies in an ADHD population or had been discontinued from future development by the sponsor (Table 1).

Table 1. Interventions for ADHD with a Serotonergic Mechanism of Action that Failed in the ADHD Indication*.

Drug Serotonin Mechanism Serotonin Activity** Highest Development Status
Amantadine 5-HT3 receptor ligand 5-HT3: IC50 = ~21–32 μM (Human)1
SERT: Ki = 100 μM (Human)2
SERT: Ki = 57–96 μM (Rat)3
• US FDA approved as adjunctive therapy for dyskinesia in PD4
Bavisant Inverse agonism associated with H3-receptor antagonists increases release of serotonin • Failed ADHD in Phase 25
Brexpiprazole 5-HT1A partial agonist
5-HT2A receptor antagonism
5-HT1A: Ki = 0.12–1.1 nM (Human)68
5-HT1A: Ki = 0.09 nM (Rat)7
5-HT1B: Ki = 365 nM (Human)8
5-HT1D: Ki = 23 nM (Human)8
5-HT1E: Ki = >10,000 nM (Human)8
5-HT2A: Ki = 0.47–3.9 nM (Human)68
5-HT2A: Ki = 3.8 nM (Rat)7
5-HT2B: Ki = 1.3–1.9 nM (Human)6,8
5-HT2C: Ki = 29 nM (Human)8
5-HT3: Ki = 754 nM (Human)8
5-HT5A: Ki = 614 nM (Human)8
5-HT6: Ki = 117 nM (Human)8
5-HT7: Ki = 3.7–6.6 nM (Human)6,8
SERT: Ki = 247 nM (Human)8
• US FDA approved as adjunctive therapy for MDD in adults, for schizophrenia in patients ⩾ 13 years, and Alzheimer’s disease dementia in adults6
• Reached phase 2 clinical trials for ADHD prior to discontinuation
Dasotraline Dopamine-norepinephrine-serotonin reuptake inhibitor SERT: IC50 = 15 nM (Human)9
SERT IC50 = 19 nM (Rat)9
• US FDA rejected NDA for ADHD10
• Development in ADHD discontinued11
Duloxetine Serotonin and norepinephrine reuptake inhibitor SERT: IC50 = 3–10 nM (Human)1217
SERT: Ki = 0.07 nM–9.3 μM (Human)1824
SERT: Ki = 0.5–8.8 nM (Rat)18,21,25,26
5-HT1A: Ki = >5000 nM (Human)21
5-HT1B: Ki = 3959 ± 810 nM (Human)21
5-HT1D: Ki = >3000 nM (Human)21
5-HT1E: Ki = 3733 ± 618 nM (Human)21
5-HT1F: Ki = 4447 ± 30 nM (Human)21
5-HT2A: Ki = 504 ± 87 nM (Human)21
5-HT2B: Ki = 2100 ± 206 nM (Human)21
5-HT2C: Ki = 916 ± 190 nM (Human)21
5-HT4: Ki = >1000 nM (Guinea Pig)21
5-HT6: Ki = 419 ± 89 nM (Human)21
5-HT7: Ki = 2261 ± 115 nM (Human)21
• US FDA approved for MDD (adults), GAD (adults/pediatric), DPNP (adults), fibromyalgia (adults/pediatric), and CMP (adults)27
• Development in ADHD discontinued
Lobeline Serotonin reuptake inhibitor SERT: Ki = >10 μM (Human)28
SERT: Ki = ~46.8 μM (Rat)29,30
5-HT2A: Ki = 7400 ± 1500 nM (Human)31
Mazindol CR Serotonin and norepinephrine reuptake inhibitor SERT: IC50 = 231 nM (Guinea Pig)2
SERT: Ki = 84–103 nM (Human)33
SERT: Ki = 49.3–272 nM (Rat)34,35
SERT: Kd = ~39 nM (Human)36
• US FDA approved for appetite suppression/obesity management but not currently available37
• Development in ADHD discontinued
• Currently being studied for narcolepsy38
Memantine 5-HT3 antagonist 5HT3: IC50 = ~2.2 μM (Human)1
5HT3A: IC50 = 2.29 ± .24 μM (Human)1
• US FDA approved for Alzheimer’s disease dementia39
• Development in ADHD discontinued
MK-0249 SERT activity
Inverse agonism associated with H3-receptor antagonists increases release of serotonin
• Failed ADHD in Phase 2a40
Modafinil SERT interactions SERT: IC50 = >232 μM (Human)41,42
SERT: IC50 = >300 μM (Rat)43
SERT: Ki = 31.3 μM (Rat)44
• US FDA approved to improve wakefulness in adult patients with excessive sleepiness associated with narcolepsy, OSA, or shift work disorder45
• US FDA did not recommend approval for children with ADHD due to safety concerns46
Molindone 5-HT2B antagonist
5-HT2A antagonist
5-HT2B: IC50 = 0.08–0.41 μM (Human)47
5-HT2A: IC50 = 14 μM (Human)47
5-HT2A: Ki = 320 nM (Human)48
5-HT2A: Ki = 501 nM (Rat)49
5-HT2A: Kd = 5800 nM (Human)50
5-HT1A: Ki = 3797 nM (Human)48
5-HT2C: Ki = >10000 nM (Human)48
5-HT6: Ki = 1008 nM (Human)48
5-HT6: Ki = >5000 nM (Rat)49
5-HT7: Ki = 265–3053 nM (Human)48,51
5-HT7: Ki = 265 ± 36 (Human)49
• Failed ADHD in Phase 352
NS2359 SERT inhibitor
Norepinephrine, dopamine, and serotonin reuptake inhibitor
SERT: IC50 = 10 nM (Human)53,54 • Development in ADHD discontinued55
• Most recently studied in cocaine-related disorders55
SEP-228432 Norepinephrine, dopamine, and serotonin reuptake inhibitor SERT: IC50 = 34 nM (Human)53
SPD473/BTS 74398 Non-selective monoamine uptake inhibitor SERT: Kd = 19 nM56
Venlafaxine Serotonin and norepinephrine reuptake inhibitor SERT: IC50 = 20–145 nM (Human)20,57
SERT: Ki = 7.8–82 nM (Human)20,21,58
SERT: Ki = 77 nM (Rat)21
SERT: ED50 = 2.0 mg/kg (Rat)21
5-HT1A: Ki = >10000 nM (Human)21
5-HT1B: Ki = >10000 nM (Human)21
5-HT1D: Ki = >10000 nM (Human)21
5-HT1E: Ki = >10000 nM (Human)21
5-HT1F: Ki = >10000 nM (Human)21
5-HT2A: Ki = 2230 nM (Human)21
5-HT2B: Ki = >10000 nM (Human)21
5-HT2C: Ki = 2004 nM (Human)21
5-HT4: Ki = >10000 nM (Guinea Pig)21
5-HT6: Ki = 2792 nM (Human)21
5-HT7: Ki = >10000 nM (Human)21
• US FDA approved for MDD, GAD, SAD, and panic disorder59
• Development in ADHD discontinued
• Follow-on compound desvenlafaxine used off-label for ADHD60
Vortioxetine 5-HT3 receptor antagonism
5-HT1A receptor agonism
5-HT7 receptor antagonism
5-HT1B partial agonist
5-HT1D antagonist
5-HT3: Ki = 3.7–23 nM (Human)61,62
5-HT3: IC50 = 12 nM (Human)62
5-HT3: IC50 = 0.18 nM (Rat)62
5-HT3: EC50 = 2100 nM (Human)62
5-HT1A: Ki = 15–39 nM (Human)6163
5-HT1A: EC50 = 200 nM (Human)62
5-HT1B: Ki = 33 nM (Human)61,62
5-HT1B: EC50 = 120 nM (Human)62
5-HT1D: Ki = 54 nM (Human)61,62
5-HT2C: Ki = 180 nM62
5-HT5A: Ki = 221 nM62
5-HT7: Ki = 19–450 nM (Human)6163
SERT: Ki = 1.6 nM (Human)61,62
SERT: IC50 = 5.4 nM (Human)61,62
SERT: IC50 = 2.9–5.3 nM (Rat)62,63
• US FDA approved for MDD61
• Failed ADHD at Phase 264

*References for this table can be found in the Supplementary Materials.

**Species has been identified as reported.

5-HT, 5-hydroxytryptamine; ADHD, attention-deficit/hyperactivity disorder; CMP, chronic musculoskeletal pain; DPNP, diabetic peripheral neuropathic pain; EC50, half maximal effective concentration; ED50, the dose of a medication that produces a specific effect in 50% of the population that has been administered that dose; GAD, generalized anxiety disorder; IC50, half-maximal inhibitory concentration; Kd, dissociation constant; Ki, inhibition constant; MDD, major depressive disorder; NDA, New Drug Application; OSA, obstructive sleep apnea; PD, Parkinson’s disease; PFC, prefrontal cortex; SAD, social anxiety disorder; SERT, serotonin transporter; US FDA, United States Food and Drug Administration.

Failed/Discontinued Compounds

Broadly, the 16 compounds that were found to have failed/been discontinued for ADHD were divided into three categories. These were (i) 5-HT receptor activity: amantadine, brexpiprazole, memantine, molindone, and vortioxetine (also has considerable serotonin transporter [SERT] inhibition); (ii) SERT activity: dasotraline, duloxetine, lobeline, mazindol CR, modafinil, NS2359, SEP-228432, SPD473/BTS 74398, and venlafaxine; and (iii) histamine H3 receptor inverse agonism, which increases serotonin release49: bavisant and MK-0249.

For those agents approved in other indications (vortioxetine, brexpiprazole, venlafaxine, duloxetine, and mazindol CR), the mechanism of action was obtained from the prescribing information. Where no prescribing information was available, the potential mechanism of action was deduced from the published scientific literature or other public domain sources.

5-HT Receptor Agonist/Antagonists

Amantadine

Although the exact mechanism of action of amantadine is unknown, it is thought to be a weak antagonist of the N-methyl-D-aspartate (NMDA) receptor.50 In addition, it has been shown in vitro to bind to 5-HT3 receptors.51

Several studies evaluating amantadine in ADHD have been published. In an open-label, uncontrolled trial of amantadine given orally to 24 patients aged 5–13 years, symptoms were evaluated at baseline and week 6 using parent and teacher ADHD rating scales (ADHD-IV rating scale).52 Treatment was titrated from 50 mg/day for 4 days to 100 mg/day for an additional 4 days, and finally to 150 mg/day. At week 6, the parent ADHD score had decreased from 41.04 ± 6.9 at baseline to 28.9 ± 8.7 (p < 0.001, effect size d = 1.5), and the teacher ADHD score from 35.8 ± 9.6 to 26.2 ± 9.5 (p < 0.001, effect size d = 1.0). Half of the patients (13/24) reported adverse events (AEs), of whom one discontinued treatment. The most common AEs were transient decrease in appetite (n = 5) and headache (n = 4).52

A randomized clinical trial in children treated with amantadine (100–150 mg/day depending on weight) or the stimulant methylphenidate (MPH) for 6 weeks showed similar improvements in ADHD symptoms with both drugs, with no significant differences between treatments on parent ADHD rating scale scores. The two types of AEs that differed significantly between treatment groups were decreased appetite (45% amantadine and 85% MPH) and restlessness (5% amantadine and 40% MPH).53 In a retrospective study of 297 pediatric patients with psychiatric symptoms, including 251 with ADHD, amantadine (⩽400 mg/day) appeared to be an effective treatment, with a success rate of 64.5% in improving symptoms.54 In addition, given that 48.1% of amantadine responders were receiving combination therapy with a stimulant (e.g., MPH or amphetamine), these data suggested that amantadine could also serve as a viable adjunct to stimulant treatment. Amantadine was also well-tolerated, with just 17/297 patients (5.7%) discontinuing due to AEs.54

Although these studies suggested that amantadine was safe and efficacious for the treatment of ADHD, no consistent dosage or regimen has been determined, and no further clinical development has been identified since 2021. This may be because the drug is available in generic forms, and no manufacturer is willing to sponsor the official clinical trial program that would be necessary to obtain regulatory approval.

Brexpiprazole

Brexpiprazole has high receptor binding affinity to norepinephrine, serotonin, and dopamine receptors. It is an antagonist at norepinephrine α1B and α2C receptors and serotonin 5-HT2A receptors, as well as partial agonist activity at serotonin 5-HT1A and dopamine D2 receptors.55,56

Data relating to the use of brexpiprazole in ADHD comes primarily from a randomized placebo-controlled phase 2 trial in 559 stimulant-naïve adults and 174 prior stimulant-nonresponding adults with ADHD, in which all participants received stimulants for 5 weeks.57 After this period, 168 stimulant-naïve patients and 68 prior stimulant nonresponders who failed treatment were randomized to receive either adjunctive brexpiprazole or placebo. When assessed using scales including the Conners’ Adult ADHD Rating Scale (CAARS), Montgomery-Asberg Depression Rating Scale, Beck Depression Inventory, Clinical Global Impression (CGI), and the Wender-Reimherr Adult Attention Deficit Disorder Scale (WRAADDS), stimulant-naïve patients were found to have no improvement with adjunctive brexpiprazole, while prior stimulant nonresponders showed a tendency toward treatment benefit only on the WRAADDS scale. AEs were similar in the brexpiprazole and placebo groups.57

Brexpiprazole is indicated for the treatment of other psychiatric disorders; however, development for the treatment of ADHD was terminated by the sponsor following phase 2 clinical study.

Memantine

Like amantadine, memantine is an NMDA receptor antagonist,58 which in vitro has been shown to have antagonistic actions at 5-HT3 receptors.51 In a randomized placebo-controlled clinical trial study of 40 adult patients (18-45 years) with ADHD, 6 weeks of treatment resulted in significant differences in behavior and attention deficit between patients treated with memantine and those receiving placebo (p < 0.001). Treatment with memantine was reported to be tolerable, with mild AEs.59

Memantine has been approved by the US Food and Drug Administration (FDA) as a treatment for Alzheimer’s disease since 2003,58 with no further clinical development in ADHD.

Molindone

Molindone is an antipsychotic medication used historically for the treatment of schizophrenia,60 although the original clinical development sponsor (Endo Pharmaceuticals, Inc.) discontinued production in 2010 due to manufacturing/sourcing difficulties.61 In vitro studies have shown that at therapeutic concentrations molindone is a potent antagonist for dopamine receptors D2S and D2L and the serotonin receptor 5-HT2B.62

Data from phase 2 studies in children with ADHD suggested that adjunctive (to MPH or amphetamine) molindone treatment was able to produce improvements in behavior, particularly symptoms of persistent impulsive aggression.63,64 A new formulation of molindone was utilized by Supernus Pharmaceuticals in the phase 3 studies CHIME 1 (NCT02618408) and 2 (NCT02618434), as well as the open-label extension study CHIME 4 (NCT02691182), for the treatment of impulsive aggression in children in conjunction with standard ADHD treatment. Although the results have not been published in a peer-reviewed journal, data published on the trial registration website indicate that the studies failed to meet the primary endpoints. Despite dropping the lower dose arm (18 mg/day) of the CHIME 1 and 2 studies after interim analyses,65 no statistical differences were observed between molindone 36 mg/day and placebo in the frequency of impulsive aggression (NCT02618408, NCT02618434, NCT02691182). AEs occurred in up to 19% of patients in the higher dose arm. A further study (NCT03597503) was planned in adolescents but was terminated due to business decisions, which were unrelated to safety. Subsequently, all clinical development activities for molindone in ADHD were halted.66

Vortioxetine

Vortioxetine is a serotonin modulator approved for major depressive disorder, which exerts its effects via 5-HT3, 5-HT1A, and 5-HT7 receptor antagonism, as well as 5-HT1B partial agonism and 5-HT1D antagonism.67,68 Vortioxetine has also shown moderate to high affinity for SERT.67

A proof-of-concept study was reported in 2019, in which 227 adult patients (18-55 years) with a diagnosis of ADHD received treatment with vortioxetine (10 or 20 mg/day) or placebo for 6 weeks. ADHD symptoms were assessed using the Adult ADHD Investigator Symptom Rating Scale (AISRS); although symptoms improved with both vortioxetine doses, there were no statistically significant differences from placebo. Both doses were well-tolerated.69 As the drug failed to meet the primary endpoint in this proof-of-concept study, no further development in ADHD was undertaken.

SERT Activity

Dasotraline

Dasotraline is a potent inhibitor of the dopamine and the norepinephrine transporter, and a weak inhibitor of SERT.70 Dasotraline was evaluated for the treatment of ADHD in an extensive clinical development program, including several late-phase placebo-controlled studies, yet failed to produce data sufficient to obtain regulatory approval.

In a placebo-controlled phase 2/3 study, treatment with dasotraline 4 mg/day was reported to significantly improve the symptoms and behaviors associated with ADHD (specifically attention and hyperactivity) in children aged 6–12 years.71 However, 2 mg/day of dasotraline did not differ significantly from placebo on the primary endpoint of ADHD Rating Scale Version IV–Home Version (RS-IV HV) score or secondary efficacy measures. Common dasotraline-related AEs included insomnia, decreased appetite, decreased weight, and irritability, and 12.2% of patients receiving dasotraline 4 mg/day discontinued treatment, compared with 1.7% in the placebo group.71 Dasotraline 4 mg/day was also found to be efficacious and generally well-tolerated in a phase 3 placebo-controlled, laboratory classroom study of children (aged 6–12) with ADHD.72 Compared with placebo, dasotraline significantly improved scores on the Swanson, Kotkin, Agler, M-Flynn, and Pelham (SKAMP)–combined score (dasotraline −3.2, placebo +2.0; p < 0.001; effect size = 0.85). Children showed improvements in the attention and deportment subscale scores. In line with the prior phase 2/3 study, the most frequent AEs associated with dasotraline were insomnia (19.6%), headache (10.7%), and decreased appetite (10.7%); 5.4% of dasotraline-treated patients discontinued the study compared with 1.8% in the placebo group.72

Dasotraline was also evaluated in a phase 3, placebo-controlled study of adults (18-55 years) with ADHD, at doses of 4 or 6 mg/day.73 However, neither dose was found to be significantly better than placebo in terms of the primary endpoint (reduction in ADHD RS-IV HV total score), although the 6-mg/day dose produced a significant effect at week 8 using unadjusted data. The most frequent AEs occurring with dasotraline (vs placebo) were insomnia, decreased appetite, dry mouth, and anxiety; these appeared to be dose-related.73

Based on these clinical trial data, the US FDA rejected a new drug application (NDA) for dasotraline in ADHD, citing the need for additional studies to more fully determine its efficacy and tolerability for this indication.74 The sponsor subsequently announced the withdrawal of the NDA and stated their intention to discontinue the clinical development program.75

Duloxetine

Duloxetine is a member of the SNRI drug class, being a potent inhibitor of neuronal serotonin and norepinephrine reuptake and a less-potent inhibitor of dopamine reuptake.76

Duloxetine is indicated for the treatment of several psychiatric disorders,76 and was evaluated in a 6-week double-blind, placebo-controlled trial in 30 adults (18–50 years) with ADHD.77 Efficacy results were mixed; duloxetine 60 mg/day had greater improvements on CGI-Severity scores (3.00 vs 4.07 for placebo; p < 0.001) and CGI-Improvement scores (2.89 vs 4.00; p < 0.001), plus greater decreases on 5 of 8 subscales of the CAARS at week 6. However, there was no effect on depression or anxiety. At week 1, 80% (12/15) of participants receiving duloxetine reported AEs, and 6 participants stopped treatment as a result.77 No additional studies of duloxetine in ADHD were identified, with no further clinical development for duloxetine in this indication.

Lobeline

Lobeline is a potent antagonist at both the α3β2 and α4β2 neuronal nicotinic receptor subtypes, and appears to perturb dopamine storage and release mechanisms.78 In vitro pharmacological evaluation has also suggested potency and selectivity for SERT inhibition.79

The effects of sublingual lobeline (7.5, 15, or 30 mg) was assessed in a proof-of-concept study using cognitive tasks and self-report measures in 9 adult volunteers (21–45 years) with ADHD. The results indicated a modest improvement in working memory but no significant improvement in attention. Patients reported not liking the taste, and nausea appeared to be a dose-related AE.80 A follow-on phase 2 clinical trial of sublingual lobeline was conducted in 13 adult patients with ADHD (NCT00664703); although the study was completed, no results have been reported to date. It is assumed that the data were not positive and that no further development was warranted.

Mazindol CR

Mazindol is an anorexiant that produces stimulatory effects on the central nervous system similar to those produced by amphetamines.81 Mazindol has been shown to alter neuronal catecholamines via its activity as an SNRI81 and was recently reported to be a partial orexin-2 receptor agonist and triple monoamine reuptake inhibitor.82

Mazindol controlled release (1–3 mg/day) was evaluated in a randomized, double-blind, placebo-controlled 6-week trial in 85 adults (aged 18–65) with ADHD.83 Based on ADHD-rating scale (ADHD-RS-DSM5) scores, mazindol improved symptoms to a greater extent than placebo (least squares mean −18.9 vs −5.7; mean difference −13.2 [95% confidence interval {CI} −18.7 to −7.6]; effect size 1.09), with differences observable from day 7. Treatment was reported to be well-tolerated, with no serious AEs, and no discontinuations due to treatment-emergent adverse events. A small effect on heart rate, similar to that produced by class II stimulants, was noted.83

Although the study authors noted that phase 3 studies were planned for mazindol in ADHD,83 according to ClinicalTrials.gov no such studies have been identified. Mazindol is currently being studied for narcolepsy (NCT05055024, NCT04923594, NCT05914194),84 and it could be assumed that the lack of clinical developmental progress in ADHD was a business decision.

Modafinil

Modafinil is a compound with wakefulness-promoting properties similar to those of amphetamines and MPH; its exact mechanism of action is unknown but appears to involve dopamine reuptake, as well as release of histamine and orexin.85,86 Data from animal studies have also indicated a dose-dependent interaction with SERT, thereby increasing dialysate serotonin levels.87

A meta-analysis of five clinical trials in a pediatric population with ADHD concluded that modafinil produced greater improvements in ADHD-RS-5 home (standardized mean difference, −0.77 [95% CI −1.11 to −0.44]) and school (−0.71 [95% CI −0.96 to −0.47]) scores compared with placebo. The most common AEs with modafinil were decreased appetite and insomnia. Discontinuation rates due to AEs were similar to the control group.88 Despite positive trial data, the US FDA advisory committee did not recommend approval of modafinil for children with ADHD because of panel members’ concern of modafinil’s potential to cause Stevens-Johnson syndrome.89

Although development of modafinil in this indication was terminated, it has been used off-label to treat ADHD.90 In addition, the follow-on drug armodafinil (an R-enantiomer of modafinil) is reported to be in use as a ‘smart drug’ to improve cognition in healthy individuals and has been used to treat ADHD off-label.91 However, studies with armodafinil have shown no significant effect on serotonin or its receptor.92

NS2359

NS2359 is a mixed monoamine reuptake inhibitor with equipotent effects on norepinephrine and dopamine transporters as well as SERT.93

In a phase 2a study in 126 adults with ADHD, no significant differences between NS2359 and placebo in terms of the primary outcome measure (reduction in investigator-rated ADHD-RS total score) were observed (7.8 vs 6.4; p < 0.45). Overall, NS2359 had no effect on ADHD symptoms, although subgroup analyses suggested a modest improvement in attention and authors noted a low dose as a potential cause. The most frequent treatment-related AE was insomnia; no serious AEs were reported.93 Further clinical development of NS2359 in ADHD was discontinued.94

SEP-228432

SEP-228432 (also called SEP-432) is a triple reuptake inhibitor of serotonin, norepinephrine, and dopamine.95 The manufacturer (Sepracor Inc.) reported an interest in developing this compound for ADHD,96 which was further reported in 2010 with the acquisition of Sepracor Inc. by Sumitomo Pharma.97

A phase 1 study in healthy volunteers (NCT01531972) was completed to ascertain the pharmacokinetics, as well as SERT and dopamine transporter (DAT) occupancy, by single-photon emission computed tomography (SPECT).98 However, by 2012, no further clinical development for SEP-228432 in ADHD had been reported.99

SPD-473

SPD-473 (also called BTS 74 398) is a mixed monoamine (serotonin, dopamine, and norepinephrine) reuptake inhibitor.100 It was reported to have completed phase 1 development in ADHD,100,101 but it appears that clinical development activities in all indications have subsequently ceased.100,101

Venlafaxine

Venlafaxine is an SNRI indicated for the treatment of several psychiatric disorders102 and has been evaluated for the treatment of ADHD in several controlled and uncontrolled clinical trials.46,103

In an open-label study of 9 adult patients (18–65 years), treatment with venlafaxine was associated with significant reductions in ADHD symptomatology at Week 2 (ADHD-RS, p < 0.02; CGI, p < 0.006), with 7/9 patients being considered responders, and only mild-to-moderate AEs being reported.104 A subsequent 6-week double-blind trial in 44 adults with ADHD failed to find a significant difference between venlafaxine and placebo in the effect on ADHD symptoms. No serious AEs were reported.105 A meta-analysis of 10 studies concluded that venlafaxine showed some efficacy in the treatment of ADHD, but was less potent than duloxetine.46 Despite the lack of official regulatory approval for this indication, venlafaxine has been reported to be used for the treatment of ADHD.106

Histamine H3 Receptor Inverse Agonism

Bavisant

An H3 receptor antagonist, bavisant was considered a good candidate for potential use in the treatment of attention and cognitive disorders107 and has been evaluated in several studies in ADHD.

In a randomized, double-blind, placebo- and active-controlled phase 2 study evaluating three dosages of bavisant (1, 3, or 10 mg/day) in adults with ADHD, the mean change in the total ADHD-RS-5 score with bavisant was −9.3 (1 mg), −11.2 (3 mg), and −12.2 (10 mg) versus −8.8 in the placebo group. For the 10-mg/day group, the change was not statistically superior to placebo (p = 0.161) and no further hierarchical testing was conducted. The highest dosage of bavisant was poorly tolerated (89.0% experienced AEs and 19.2% discontinued as a result).107 Three early-stage studies in children (NCT00890240 and NCT00890292) and adults (NCT00566449) with ADHD were also completed, but to date, no results have been published.

MK-0249

MK-0249 was a developmental compound investigated in various cognitive defects, including ADHD,108 sleepiness associated with obstructive sleep apnea,109 and Alzheimer’s disease.110 MK-0249 administration acts on the H3 receptor to increase histamine release,108 which is pro-cognitive and could assist with ADHD symptoms; the H3 receptor also acts as a heteroreceptor that regulates the release of neurotransmitters such as serotonin.108

In a randomized, double-blind, placebo-controlled, crossover phase 2a study of MK-0249 (5-10 mg/day) versus MPH, MK-0249 did not differ from placebo (p = 0.341) in terms of AISRS scores, whereas MPH produced significant improvements (p < 0.001). Although MK-0249 was reported to be generally well-tolerated, 5/37 (13.5%) patients discontinued treatment due to AEs, compared with 6.8% in the MPH group and 3.7% in the placebo groups. A greater percentage of patients reported insomnia with MK-0249 treatment compared with placebo (32% vs 11%, respectively).108 Given the unfavorable outcome in this study, plus failures in the studies for obstructive sleep apnea109 and Alzheimer’s disease,110 further clinical development of MK-0249 was discontinued.

Discussion

Evaluating the Developmental Failures

Due to the large body of evidence indicating a role for 5-HT in mood disorders,2,3 and their very high comorbidity in patients with ADHD, many pharmaceutical companies have looked at serotonergic compounds for the treatment of ADHD, with mixed success. For every drug that has been approved in this field, several others have either failed clinical trials or have been withdrawn from development. Some compounds were assessed in a clinical trial program involving multiple randomized studies yet still failed to generate data that were sufficiently robust to obtain regulatory approval. Other compounds, despite showing promise in early-stage trials, were not investigated further, possibly because there was no sponsor willing to fund the trials likely needed to progress to an NDA or for other business-related reasons. The reasons for discontinuation or non-approval are multifactorial, with study design factors such as patient selection, ADHD diagnostic criteria, and scales and endpoints each playing a part. However, it is also possible that patient selection and appropriate dose selection, including the need to balance effectiveness with safety may underlie at least some of the problems observed during drug development for ADHD.

Potency and selectivity are key attributes when considering efficacy and tolerability. Off-target effects resulting in AEs may reduce the dosage range for a particular compound, thereby decreasing the ability of the prescribing physician to maximize its effectiveness sufficiently to control symptoms. As shown in Table 1, the reported half-maximal inhibitory concentrations (IC50) and inhibition constants (Ki) vary widely among the drugs described here, suggesting that each will have a different benefit-risk profile during ADHD treatment.

Moreover, specific serotonergic efficacy and tolerability effects are likely to be further complicated by AEs elicited in response to the other effects of a given drug, e.g., toxicity driven by binding to/inhibition of noradrenaline and dopamine receptors and transporters. However, this level of information would require a much deeper investigation of the exact mechanism of action than is available for many approved and failed ADHD treatment candidates, several of which remain imperfectly understood. It is likely that there is a ‘Goldilocks zone’, in which the effects in the brain are sufficient to manage ADHD symptoms without triggering the type and severity of AEs likely to lead to discontinuation. In support of this, we note that several studies of failed drugs (including amantadine, molindone, dasotraline, and bavisant) struggled to determine an optimal dose, and some approved drugs (notably amphetamines and MPH) are routinely titrated by physicians in an attempt to obtain the best outcomes. Patterns of drug administration (for short durations or intermittently) may also limit the therapeutic value of developmental serotonergic drugs.111 The fact of a particular compound having a known serotonin effect and proven efficacy in other neurobehavioral disorders may not be enough to indicate that it should be tested for effectiveness in ADHD. Instead, a deeper understanding of its potency and selectivity appears necessary before embarking on clinical trials.

Current Treatment Landscape of Serotonergic Drugs Used in ADHD

Several of the best-known and most commonly used pharmacological treatments for ADHD are associated with effects on serotonergic neurotransmission. Amphetamines are non-catecholamine, sympathomimetic amines,112114 but have also been shown to have affinity for SERT, thereby increasing extracellular serotonin.115,116 Amphetamines have been shown to robustly improve the severity of ADHD symptoms but are associated with AEs (e.g., elevated blood pressure, cardiovascular conditions, gastrointestinal symptoms, decreased appetite, long-term suppression of growth, and neuropsychiatric symptoms, such as irritability, mania, and psychosis117,118) that can result in high discontinuation rates.119

The mechanism of action of MPH in ADHD is not fully understood, but it is thought to block dopamine and norepinephrine reuptake.120,121 The evidence for a serotonergic role is not as conclusive as for amphetamines; the affinity of MPH for SERT is lower than for dopamine or norepinephrine transporters,122 although MPH has been reported to have agonist activity at the 5-HT1A receptor.123 Some researchers have found no indication of an MPH effect on 5-HT pathways,115 while others have suggested that MPH administration can rebalance serotonin levels in the brain and blood, thereby reducing hyperactivity124,125 and synchronizing biological rhythms126 in patients with ADHD. The relative activity on the serotonin system for a given patient could be subject to inter-individual variability. MPH is widely used in ADHD, although studies have shown that dosing and clinical response are variable,127 likely due to genetic differences in the activity of the carboxylesterase 1 enzyme, which metabolizes MPH,128 and as a result of titration in an attempt to minimize AEs.129

Atomoxetine is reported to be a selective NRI130 but has also been shown to bind in vitro to SERT.131 The effectiveness of atomoxetine was established following an extensive development program involving multiple randomized controlled clinical studies in both children (4 studies) and adults (2 studies).132,133 Viloxazine ER, another selective NRI,134 has also been shown to increase brain serotonin levels via its actions at 5-HT receptors (5-HT2B and 5-HT7 antagonist and 5-HT2C agonist).135 It can be hypothesized that the 5-HT2C agonist activity of viloxazine regulates dopamine signaling in both the midbrain and striatal regions via interactions with gamma-aminobutyric acid interneurons,136139 which have been shown to reduce impulsive behavior.140

In addition, there are several drugs which have some degree of serotonergic activity that are not approved by the US FDA for the treatment of ADHD but are nonetheless reported to be used for this purpose (often concomitantly alongside stimulants). Agomelatine is a melatonin agonist and selective serotonin antagonist, with effects at 5-HT2C and 5-HT2B receptors.141143 It is currently approved in other countries, but not in the US, for the treatment of adult depression,141 and has been reported to have psychostimulant activities, with the ability to regulate cognitive activity and circadian rhythms.143 In a small clinical trial in ADHD, the addition of agomelatine to MPH or atomoxetine suggested clinical efficacy, particularly in terms of the impact on comorbid sleep, anxiety, or oppositional defiance disorder.144 In another study, agomelatine monotherapy was found to be as effective as MPH on parent and teacher rating scale scores.145 Across all published studies and indications, agomelatine is considered well-tolerated with high levels of adherence.143

The TCA desipramine is indicated for the treatment of depression in the US146 and has been reported to be used in the treatment of ADHD.147,148 It appears to exert its effects via blocking norepinephrine and serotonin reuptake,148 and has been reported to alter 5-HT receptor sensitivity.149 However, although clinical studies in ADHD have indicated that desipramine can decrease clinical symptoms, there are serious concerns regarding its safety, particularly in children.150 Imipramine is another TCA indicated in the US for depression151 and has been linked to the treatment of ADHD,152 although the clinical data supporting its use are older.45 It exerts a serotonergic effect via inhibition of SERT and alterations in 5-HT1A receptor sensitivity.153,154

Compounds Currently in Development

Several compounds with some degree of serotonergic activity are currently in clinical development for the treatment of ADHD. It will be of future interest to assess the final regulatory outcome in the light of what is known about the serotonergic potency and selectivity of each of these drugs.

Cariprazine is an atypical antipsychotic indicated by the US FDA for several psychiatric disorders.155 It acts as a partial agonist at the dopamine D2 and D3 receptors and serotonin 5-HT1A receptors, and as an antagonist at 5-HT2A and 5-HT2B receptors. Its binding affinity is reported to be high at 5-HT2B receptors (Ki = 0.58 nM) and 5-HT1A receptors (Ki = 2.6 nM), moderate at 5-HT2A receptors (Ki = 18.8 nM), and low at 5-HT2C receptors (Ki = 155 nM).155 A flexible-dose clinical trial in adults with ADHD is currently recruiting (NCT04843423).

Centanafadine is a first-in-class norepinephrine, dopamine, and serotonin reuptake inhibitor (NDSRI) being developed for the treatment of ADHD in children, adolescents, and adults.156 It has IC50 values of 6 nM for norepinephrine, 38 nM for dopamine, and 83 nM for serotonin transporter reuptake.157 The NDSRI mechanism may provide a more favorable safety and tolerability profile when compared to the current standard of care. Moreover, the serotonergic activity of centanafadine has been postulated to mitigate treatment-related AEs such as sleep disturbances and changes in appetite.157 Modulation of serotonin could also assist with associated symptoms of ADHD that are not as well-treated by the current standard of care, including anxiety and depression.157 To date, the sponsor has completed 9 clinical studies in ADHD, with 2 currently ongoing (NCT05279313 and NCT05428033). Data from two phase 3 studies of the sustained-release formulation conducted in adults with ADHD reported statistically significant improvements in AISRS total scores versus placebo and a low rate of treatment-emergent AEs leading to discontinuation.158 Two phase 3 studies in pediatric ADHD patients have also been completed, but only topline results have been reported to date; both trials met the primary efficacy endpoint for the higher dose studied, with no new safety concerns identified.156

Solriamfetol, a dopamine and norepinephrine reuptake inhibitor (DNRI), is indicated to improve wakefulness in adults with excessive daytime sleepiness that is associated with narcolepsy or obstructive sleep apnea.159 Preclinical data suggest that solriamfetol exhibits agonist activity at the 5-HT1A receptor160 as well as trace amine-associated receptor 1 (TAAR1) agonism,160 which is believed to have several effects on serotonin neurotransmission, including enhancing the potency of 5-HT1A agonists.161 Agonist activity at TAAR1 was also shown to be involved in serotonergic neurotransmission.162,163 According to the manufacturer, solriamfetol has low binding affinity for the serotonin transporter (Ki = 81.5 μM) and does not inhibit serotonin reuptake (IC50 > 100 μM).159 Following the publication of positive data from a dose-optimization pilot study of solriamfetol in 60 adults with ADHD,164 a phase 3 clinical study was initiated and is currently recruiting (NCT05972044).

Clinical Implications and Future Treatment Expectations

Given that ADHD is one of the most common neuropsychiatric disorders,1113,165 and places a high burden on individuals and society, there is a great deal of interest in developing safe and effective treatments. Currently approved therapies include stimulant drugs (MPH and amphetamine derivatives) and a handful of non-stimulants, but concerns relating to efficacy and safety remain.3437

The unfavorable outcome rate for drugs intended for ADHD treatment is large, and the bar for evidence robust enough for regulatory approval is set high.101 Nonetheless, there has been considerable progress in the understanding of ADHD neurobiology, and each failure expands our knowledge base and should enable more rational drug development, improved trial procedures, and better optimized dosing in the future.

While the initial focus of drug development was on the catecholamines, norepinephrine and dopamine,101,166 there is also a large body of evidence indicating a role for the serotonergic system in ADHD. Given the large number of failed trials, it does not appear that serotonergic modulation alone is likely to produce efficacious treatments for ADHD. Instead, it seems likely that altering the balance between monoamine neurotransmission systems is likely to provide the best outcomes, with serotonergic activity complementing effects on norepinephrine and dopamine. Notably, one of the drugs currently in development, centanafadine, is an NDSRI targeting all three of these neurotransmitter systems; based on clinical trial data to date, centanafadine demonstrated clinically meaningful efficacy with a favorable safety and tolerability profile. The use of this and other multimodal agents is expected to expand the pool of available drugs for ADHD, simultaneously providing robust and consistent efficacy alongside a favorable safety profile and lower potential for abuse, and delivering additional benefits with regard to comorbidities (such as sleep, anxiety, and mood).166

Limitations of this Review

This narrative review does not represent an exhaustive analysis of every compound with serotonergic activity ever tested in ADHD. Instead, it is intended to provide a summary of this often-overlooked area of research, to consider the role of 5-HT in the pathology and treatment of ADHD, and to generate discussion regarding the high unfavorable outcome rate to date. As this study was reliant on information available in the public domain, several compounds for which no data have been made publicly available have been omitted and some supporting sources that were not peer-reviewed have been included.

Conclusions

There is an ever-increasing body of evidence suggesting a role for 5-HT in the neurobiology of ADHD and indicating that modulation of serotonergic neurotransmission and rebalancing of monoamines within the brain may provide an avenue of drug development by which outcomes for patients with ADHD can be improved. In this evidence-based narrative review, many compounds with serotonergic activity that have been studied in ADHD were identified, and pinpointed key reasons for a halt in the developmental process, which include finding the correct dose and selectivity to produce a balance between potency and tolerability. At the current time, clinical studies of several drug candidates with serotonergic activity are ongoing, and it is anticipated that such treatments may positively impact multiple ADHD-associated symptoms, including anxiety and emotional dysregulation, in addition to inattention and hyperactivity.

Acknowledgments

The authors thank Sally-Anne Mitchell, PhD, and Julia L. Jones, PhD, of The Medicine Group for providing medical writing support, which was funded by Otsuka Pharmaceutical Co., Ltd., and in accordance with Good Publication Practice 2022 guidelines (https://www.ismpp.org/gpp-2022). The authors also thank Olesya Taylor for her insight and contributions to this work.

Footnotes

Disclosures

Craig Chepke has served on the advisory boards of AbbVie, Acadia, Alkermes, Axsome, Biogen, Bristol Myers Squibb, Corium, Idorsia, Intra-Cellular, Janssen, Lundbeck, Moderna, Neurocrine, Noven, Otsuka, Sage, Sumitomo, and Teva; Advisory Board (spouse): Bristol Myers Squibb, Otsuka; served as a consultant for AbbVie, Acadia, Alkermes, Axsome, Biogen, Boehringer Ingelheim, Corium, Intra-Cellular, Janssen, Lundbeck, MedinCell, Moderna, Neurocrine, Noven, Otsuka, Sage, Sumitomo, Supernus, and Teva; has received grant or research support from Acadia, Axsome, Harmony, Neurocrine, and Teva; and has been on the speaker’s bureau for AbbVie, Acadia, Alkermes, Axsome, Bristol Myers Squibb, Corium, Intra-Cellular, Janssen, Lundbeck, Merck, Neurocrine, Noven, Otsuka, Sumitomo, and Teva. Elizabeth Brunner is a full-time employee of Otsuka Pharmaceutical Development & Commercialization, Inc. Andrew J. Cutler has been a consultant or advisory board member for AbbVie, Acadia, Alfasigma, Alkermes, Anavex Life Sciences, Axsome, Biogen, Biohaven, BioXcel, Boehringer Ingelheim, Brii Biosciences, Bristol Myers Squibb, Cerevel, Chase Therapeutics, Cognitive Research, Corium, Delpor, Evolution Research Group, 4M Therapeutics, Intra-Cellular Therapies, Ironshore Pharmaceuticals, Janssen/J&J, Jazz Pharma, Karuna, LivoNova, Lundbeck, Luye Pharma, Maplight Therapeutics, MedAvante-ProPhase, Mentavi, Neumora, Neurocrine, Neuroscience Education Institute, NeuroSigma, Noven, Otsuka, PaxMedica, PureTech Health, Relmada, Reviva, Sage Therapeutics, Sumitomo Pharma America, Sunovion, Supernus, Teva, Thynk, Tris Pharma, Vanda Pharmaceuticals, VistaGen, and VivoSense; has been on a speaker bureau for AbbVie, Acadia, Alfasigma, Alkermes, Axsome, BioXcel, Corium, Intra-Cellular Therapies, Ironshore Pharmaceuticals, Janssen/J&J, Lundbeck, Neurocrine, Noven, Otsuka, Sunovion, Supernus, Teva, Tris Pharma, and Vanda Pharmaceuticals; and has stock options in 4M Therapeutics and Relmada.

Contributor Information

Craig Chepke, Chepke, MD, DFAPA, Excel Psychiatric Associates, Huntersville, NC; Atrium Health, Charlotte, NC..

Elizabeth Brunner, Brunner, MD, Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ..

Andrew J Cutler, Cutler, MD, SUNY Upstate Medical University, Lakewood Ranch, FL..

Supplementary Material

References

  • 1.Kanova M, Kohout P. Serotonin—Its synthesis and roles in the healthy and the critically ill. Int J Mol Sci . 2021;22(9):4837. doi: 10.3390/ijms22094837. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lin SH, Lee LT, Yang YK. Serotonin and mental disorders: A concise review on molecular neuroimaging evidence. Clin Psychopharmacol Neurosci . 2014;12(3):196–202. doi: 10.9758/cpn.2014.12.3.196. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Pourhamzeh M, Moravej FG, Arabi M et al. The roles of serotonin in neuropsychiatric disorders. Cell Mol Neurobiol . 2022;42(6):1671–1692. doi: 10.1007/s10571-021-01064-9. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Lucki I. The spectrum of behaviors influenced by serotonin. Biol Psychiatry . 1998;44(3):151–162. doi: 10.1016/s0006-3223(98)00139-5. doi: [DOI] [PubMed] [Google Scholar]
  • 5.Hou YW, Xiong P, Gu X, Huang X, Wang M, Wu J. Association of serotonin receptors with attention deficit hyperactivity disorder: A systematic review and meta-analysis. Curr Med Sci . 2018;38(3):538–551. doi: 10.1007/s11596-018-1912-3. doi: [DOI] [PubMed] [Google Scholar]
  • 6.Lin J, Liu W, Guan J et al. Latest updates on the serotonergic system in depression and anxiety. Front Synaptic Neurosci . 2023;15:1124112. doi: 10.3389/fnsyn.2023.1124112. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hirsch O, Chavanon M, Riechmann E, Christiansen H. Emotional dysregulation is a primary symptom in adult attention-deficit/hyperactivity disorder (ADHD) J Affect Disord . 2018;232:41–47. doi: 10.1016/j.jad.2018.02.007. doi: [DOI] [PubMed] [Google Scholar]
  • 8.Shaw P, Stringaris A, Nigg J, Leibenluft E. Emotion dysregulation in attention deficit hyperactivity disorder. Am J Psychiatry . 2014;171(3):276–293. doi: 10.1176/appi.ajp.2013.13070966. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Faraone SV, Banaschewski T, Coghill D et al. The World Federation of ADHD International Consensus Statement: 208 evidence-based conclusions about the disorder. Neurosci Biobehav Rev . 2021;128:789–818. doi: 10.1016/j.neubiorev.2021.01.022. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.GBD 2019 Mental Disorders Collaborators. Global, regional, and national burden of 12 mental disorders in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Psychiatry . 2022;9(2):137–150. doi: 10.1016/S2215-0366(21)00395-3. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ayano G, Demelash S, Gizachew Y, Tsegay L, Alati R. The global prevalence of attention deficit hyperactivity disorder in children and adolescents: An umbrella review of meta-analyses. J Affect Disord . 2023;339:860–866. doi: 10.1016/j.jad.2023.07.071. doi: [DOI] [PubMed] [Google Scholar]
  • 12.Salari N, Ghasemi H, Abdoli N et al. The global prevalence of ADHD in children and adolescents: A systematic review and meta-analysis. Ital J Pediatr . 2023;49(1):48. doi: 10.1186/s13052-023-01456-1. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Song P, Zha M, Yang Q, Zhang Y, Li X, Rudan I. The prevalence of adult attention-deficit hyperactivity disorder: A global systematic review and meta-analysis. J Glob Health . 2021;11:04009. doi: 10.7189/jogh.11.04009. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cortese S, Song M, Farhat LC et al. Incidence, prevalence, and global burden of ADHD from 1990 to 2019 across 204 countries: Data, with critical re-analysis, from the Global Burden of Disease study. Mol Psychiatry . 2023;28(11):4823–4830. doi: 10.1038/s41380-023-02228-3. doi: [DOI] [PubMed] [Google Scholar]
  • 15.Li Y, Yan X, Li Q et al. Prevalence and trends in diagnosed ADHD among US children and adolescents, 2017–2022. JAMA Netw Open . 2023;6(10):e2336872. doi: 10.1001/jamanetworkopen.2023.36872. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Abdelnour E, Jansen MO, Gold JA. ADHD diagnostic trends: Increased recognition or overdiagnosis? . Mo Med . 2022;119(5):467–473. [PMC free article] [PubMed] [Google Scholar]
  • 17.London AS, Landes SD. Cohort change in the prevalence of ADHD among U.S. adults: Evidence of a gender-specific historical period effect. J Atten Disord . 2021;25(6):771–782. doi: 10.1177/1087054719855689. doi: [DOI] [PubMed] [Google Scholar]
  • 18.Sibley MH, Arnold LE, Swanson JM et al. Variable patterns of remission from ADHD in the multimodal treatment study of ADHD. Am J Psychiatry . 2022;179(2):142–151. doi: 10.1176/appi.ajp.2021.21010032. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gascon A, Gamache D, St-Laurent D, Stipanicic A. Do we over-diagnose ADHD in North America? A critical review and clinical recommendations. J Clin Psychol . 2022;78(12):2363–2380. doi: 10.1002/jclp.23348. doi: [DOI] [PubMed] [Google Scholar]
  • 20.Berchiatti M, Ferrer A, Badenes-Ribera L, Longobardi C. School adjustments in children with attention deficit hyperactivity disorder (ADHD): Peer relationships, the quality of the student-teacher relationship, and children’s academic and behavioral competencies. Journal of Applied School Psychology . 2022;38(3):241–261. doi: 10.1080/15377903.2021.1941471. doi: [DOI] [Google Scholar]
  • 21.Pawaskar M, Fridman M, Grebla R, Madhoo M. Comparison of quality of life, productivity, functioning and self-esteem in adults diagnosed with ADHD and with symptomatic ADHD. J Atten Disord . 2020;24(1):136–144. doi: 10.1177/1087054719841129. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Varrasi S, Boccaccio FM, Guerrera CS, Platania GA, Pirrone C, Castellano S. Schooling and occupational outcomes in adults with ADHD: Predictors of success and support strategies for effective learning. Educ Sci . 2023;13(1):37. doi: 10.3390/educsci13010037. doi: [DOI] [Google Scholar]
  • 23.Choi WS, Woo YS, Wang SM, Lim HK, Bahk WM. The prevalence of psychiatric comorbidities in adult ADHD compared with non-ADHD populations: A systematic literature review. PLoS One . 2022;17(11):e0277175. doi: 10.1371/journal.pone.0277175. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Van de Glind G, Brynte C, Skutle A. The International Collaboration on ADHD and Substance Abuse (ICASA): Mission, results, and future activities. Eur Addict Res . 2020;26(4–5):173–178. doi: 10.1159/000508870. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Brunkhorst-Kanaan N, Libutzki B, Reif A, Larsson H, McNeill RV, Kittel-Schneider S. ADHD and accidents over the life span - A systematic review. Neurosci Biobehav Rev . 2021;125:582–591. doi: 10.1016/j.neubiorev.2021.02.002. doi: [DOI] [PubMed] [Google Scholar]
  • 26.Dalsgaard S, Ostergaard SD, Leckman JF, Mortensen PB, Pedersen MG. Mortality in children, adolescents, and adults with attention deficit hyperactivity disorder: A nationwide cohort study. Lancet . 2015;385(9983):2190–2196. doi: 10.1016/S0140-6736(14)61684-6. doi: [DOI] [PubMed] [Google Scholar]
  • 27.Nunez-Jaramillo L, Herrera-Solis A, Herrera-Morales WV. ADHD: Reviewing the causes and evaluating solutions. J Pers Med . 2021;11(3):166. doi: 10.3390/jpm11030166. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Grimm O, Kranz TM, Reif A. Genetics of ADHD: What should the clinician know? Curr Psychiatry Rep . 2020;22(4):18. doi: 10.1007/s11920-020-1141-x. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Curatolo P, D’Agati E, Moavero R. The neurobiological basis of ADHD. Ital J Pediatr . 2010;36(1):79. doi: 10.1186/1824-7288-36-79. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.van Gaalen MM, van Koten R, Schoffelmeer AN, Vanderschuren LJ. Critical involvement of dopaminergic neurotransmission in impulsive decision making. Biol Psychiatry . 2006;60(1):66–73. doi: 10.1016/j.biopsych.2005.06.005. doi: [DOI] [PubMed] [Google Scholar]
  • 31.Franca TFA, Pompeia S. Reappraising the role of dopamine in adolescent risk-taking behavior. Neurosci Biobehav Rev . 2023;147:105085. doi: 10.1016/j.neubiorev.2023.105085. doi: [DOI] [PubMed] [Google Scholar]
  • 32.Zhang Y, Chen Y, Xin Y, Peng B, Liu S. Norepinephrine system at the interface of attention and reward. Prog Neuropsychopharmacol Biol Psychiatry . 2023;125:110751. doi: 10.1016/j.pnpbp.2023.110751. doi: [DOI] [PubMed] [Google Scholar]
  • 33.Stuhec M, Munda B, Svab V, Locatelli I. Comparative efficacy and acceptability of atomoxetine, lisdexamfetamine, bupropion and methylphenidate in treatment of attention deficit hyperactivity disorder in children and adolescents: A meta-analysis with focus on bupropion. J Affect Disord . 2015;178:149–159. doi: 10.1016/j.jad.2015.03.006. doi: [DOI] [PubMed] [Google Scholar]
  • 34.Spencer T, Biederman J, Wilens T et al. A large, double-blind, randomized clinical trial of methylphenidate in the treatment of adults with attention-deficit/hyperactivity disorder. Biol Psychiatry . 2005;57(5):456–463. doi: 10.1016/j.biopsych.2004.11.043. doi: [DOI] [PubMed] [Google Scholar]
  • 35.Spencer T, Biederman J, Wilens T, Harding M, O’Donnell D, Griffin S. Pharmacotherapy of attention-deficit hyperactivity disorder across the life cycle. J Am Acad Child Adolesc Psychiatry . 1996;35(4):409–432. doi: 10.1097/00004583-199604000-00008. doi: [DOI] [PubMed] [Google Scholar]
  • 36.Childress AC, Sallee FR. Attention-deficit/hyperactivity disorder with inadequate response to stimulants: approaches to management. CNS Drugs . 2014;28(2):121–129. doi: 10.1007/s40263-013-0130-6. doi: [DOI] [PubMed] [Google Scholar]
  • 37.Schneider BN, Enenbach M. Managing the risks of ADHD treatments. Curr Psychiatry Rep . 2014;16(10):479. doi: 10.1007/s11920-014-0479-3. doi: [DOI] [PubMed] [Google Scholar]
  • 38.Oades RD. Role of the serotonin system in ADHD: Treatment implications. Expert Rev Neurother . 2007 Oct;7(10):1357–1374. doi: 10.1586/14737175.7.10.1357. doi: [DOI] [PubMed] [Google Scholar]
  • 39.Vanicek T, Kutzelnigg A, Philippe C et al. Altered interregional molecular associations of the serotonin transporter in attention deficit/hyperactivity disorder assessed with PET. Hum Brain Mapp . 2017;38(2):792–802. doi: 10.1002/hbm.23418. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Banerjee E, Nandagopal K. Does serotonin deficit mediate susceptibility to ADHD? Neurochem Int . 2015;82:52–68. doi: 10.1016/j.neuint.2015.02.001. doi: [DOI] [PubMed] [Google Scholar]
  • 41.Ruocco LA, Treno C, Gironi Carnevale UA et al. Prepuberal stimulation of 5-HT7-R by LP-211 in a rat model of hyper-activity and attention-deficit: Permanent effects on attention, brain amino acids and synaptic markers in the fronto-striatal interface. PLoS One . 2014;9(4):e83003. doi: 10.1371/journal.pone.0083003. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Sharp T, Barnes NM. Central 5-HT receptors and their function; Present and future. Neuropharmacology . 2020;177:108155. doi: 10.1016/j.neuropharm.2020.108155. doi: [DOI] [PubMed] [Google Scholar]
  • 43.Dunn GA, Nigg JT, Sullivan EL. Neuroinflammation as a risk factor for attention deficit hyperactivity disorder. Pharmacol Biochem Behav . 2019;182:22–34. doi: 10.1016/j.pbb.2019.05.005. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Park P, Caballero J, Omidian H. Use of serotonin norepinephrine reuptake inhibitors in the treatment of attention-deficit hyperactivity disorder in pediatrics. Ann Pharmacother . 2014;48(1):86–92. doi: 10.1177/1060028013506561. doi: [DOI] [PubMed] [Google Scholar]
  • 45.Otasowie J, Castells X, Ehimare UP, Smith CH. Tricyclic antidepressants for attention deficit hyperactivity disorder (ADHD) in children and adolescents. Cochrane Database Syst Rev . 2014;(9):CD006997. doi: 10.1002/14651858.CD006997.pub2. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Dezfouli RA, Hosseinpour A, Ketabforoush S, Daneshzad E. Efficacy, safety, and tolerability of serotonin-norepinephrine reuptake inhibitors in controlling ADHD symptoms: A systematic review and meta-analysis. Middle East Current Psychiatry . 2024;31(1):8. doi: 10.1186/s43045-024-00400-1. doi: [DOI] [Google Scholar]
  • 47.Barrickman L, Noyes R, Kuperman S, Schumacher E, Verda M. Treatment of ADHD with fluoxetine: A preliminary trial. J Am Acad Child Adolesc Psychiatry . 1991;30(5):762–767. [PubMed] [Google Scholar]
  • 48.Padilha S, Virtuoso S, Tonin FS, Borba HHL, Pontarolo R. Efficacy and safety of drugs for attention deficit hyperactivity disorder in children and adolescents: a network meta-analysis. Eur Child Adolesc Psychiatry . 2018;27(10):1335–1345. doi: 10.1007/s00787-018-1125-0. doi: [DOI] [PubMed] [Google Scholar]
  • 49.Esbenshade TA, Browman KE, Bitner RS, Strakhova M, Cowart MD, Brioni JD. The histamine H3 receptor: An attractive target for the treatment of cognitive disorders. Br J Pharmacol . 2008;154(6):1166–81. doi: 10.1038/bjp.2008.147. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. GOCOVRI (amantadine) extended release capsules, for oral use [prescribing information]. Revised 8/2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/208944lbl.pdf. [Google Scholar]
  • 51.Rammes G, Rupprecht R, Ferrari U, Zieglgansberger W, Parsons CG. The N-methyl-D-aspartate receptor channel blockers memantine, MRZ 2/579 and other amino-alkyl-cyclohexanes antagonise 5-HT(3) receptor currents in cultured HEK-293 and N1E-115 cell systems in a non-competitive manner. Neurosci Lett . 2001;306(1–2):81–84. doi: 10.1016/s0304-3940(01)01872-9. doi: [DOI] [PubMed] [Google Scholar]
  • 52.Donfrancesco R, Calderoni D, Vitiello B. Open-label amantadine in children with attention-deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol . 2007;17(5):657–664. doi: 10.1089/cap.2006.0128. doi: [DOI] [PubMed] [Google Scholar]
  • 53.Mohammadi MR, Kazemi MR, Zia E, Rezazadeh SA, Tabrizi M, Akhondzadeh S. Amantadine versus methylphenidate in children and adolescents with attention deficit/hyperactivity disorder: A randomized, double-blind trial. Hum Psychopharmacol . 2010;25(7–8):560–565. doi: 10.1002/hup.1154. doi: [DOI] [PubMed] [Google Scholar]
  • 54.Morrow K, Choi S, Young K, Haidar M, Boduch C, Bourgeois JA. Amantadine for the treatment of childhood and adolescent psychiatric symptoms. Proc (Bayl Univ Med Cent) . 2021;34(5):566–570. doi: 10.1080/08998280.2021.1925827. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Maeda K, Sugino H, Akazawa H et al. Brexpiprazole I: in vitro and in vivo characterization of a novel serotonin-dopamine activity modulator. J Pharmacol Exp Ther . 2014;350(3):589–604. doi: 10.1124/jpet.114.213793. doi: [DOI] [PubMed] [Google Scholar]
  • 56. REXULTI® (brexpiprazole) tablets, for oral use [prescribing information]. Revised 5/2023. https://www.otsuka-us.com/sites/g/files/qhldwo8366/files/media/static/Rexulti-PI.pdf. [Google Scholar]
  • 57.Reimherr FW, Gift TE, Steans TA et al. The use of brexpiprazole combined with a stimulant in adults with treatment-resistant attention-deficit/hyperactivity disorder. J Clin Psychopharmacol . 2022;42(5):445–453. doi: 10.1097/JCP.0000000000001592. doi: [DOI] [PubMed] [Google Scholar]
  • 58. NAMENDA® (memantine hydrochloride) tablets, for oral use [prescribing information]. Revised 11/2018. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/021487s025lbl.pdf. [Google Scholar]
  • 59.Mohammadzadeh S, Ahangari TK, Yousefi F. The effect of memantine in adult patients with attention deficit hyperactivity disorder. Hum Psychopharmacol . 2019;34(1):e2687. doi: 10.1002/hup.2687. doi: [DOI] [PubMed] [Google Scholar]
  • 60. MOBAN® (molindone hydrochloride tablets, USP) [prescribing information]. Revised 6/2009. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/017111s066lbl.pdf. [Google Scholar]
  • 61.US Food and Drug Administration. Determination that MOBAN (Molindone Hydrochloride) Tablets (5 milligrams, 10 milligrams, 25 milligrams, 50 milligrams, and 100 milligrams) and capsules (5 milligrams, 10 milligrams, and 25 milligrams) were not withdrawn from sale for reasons of safety or effectiveness. Federal Register . 2013;78(215):66742–66743. [Google Scholar]
  • 62.Yu C, Gopalakrishnan G. In vitro pharmacological characterization of SPN-810M (molindone) J Exp Pharmacol . 2018;10:65–73. doi: 10.2147/JEP.S180777. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Stocks JD, Taneja BK, Baroldi P, Findling RL. A phase 2a randomized, parallel group, dose-ranging study of molindone in children with attention-deficit/hyperactivity disorder and persistent, serious conduct problems. J Child Adolesc Psychopharmacol . 2012;22(2):102–11. doi: 10.1089/cap.2011.0087. doi: [DOI] [PubMed] [Google Scholar]
  • 64.Ceresoli-Borroni G, Nasser A, Adewole T et al. A double-blind, randomized study of extended-release molindone for impulsive aggression in ADHD. J Atten Disord . 2021;25(11):1564–1577. doi: 10.1177/1087054720909084. doi: [DOI] [PubMed] [Google Scholar]
  • 65.Supernus Pharmaceuticals. News Release Details September 18, 2017. https://ir.supernus.com/news-releases/news-release-details/supernus-provides-update-spn-810-phase-iii-clinical-trials-0?ReleaseID=1040834. [Google Scholar]
  • 66.Surran C. Supernus Pharma’s SPN-810 trial did not meet primary endpoint. [June 11, 2024]; Seeking Alpha. Accessed. https://seekingalpha.com/news/3545617-supernus-pharmas-spnminus-810-trial-not-meet-primary-endpoint . [Google Scholar]
  • 67. TRINTELLIX (vortioxetine) tablets, for oral use [prescribing information]. Revised 8/2023. https://content.takeda.com/?contenttype=PI&product=TRI&language=ENG&country=GBL&documentnumber=1. [Google Scholar]
  • 68.D’Agostino A, English CD, Rey JA. Vortioxetine (brintellix): A new serotonergic antidepressant. P T . 2015;40(1):36–40. [PMC free article] [PubMed] [Google Scholar]
  • 69.Biederman J, Lindsten A, Sluth LB et al. Vortioxetine for attention deficit hyperactivity disorder in adults: A randomized, double-blind, placebo-controlled, proof-of-concept study. J Psychopharmacol . 2019;33(4):511–521. doi: 10.1177/0269881119832538. doi: [DOI] [PubMed] [Google Scholar]
  • 70.Hopkins SC, Sunkaraneni S, Skende E et al. Pharmacokinetics and exposure-response relationships of dasotraline in the treatment of attention-deficit/hyperactivity disorder in adults. Clin Drug Investig . 2016;36(2):137–146. doi: 10.1007/s40261-015-0358-7. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Findling RL, Adler LA, Spencer TJ et al. Dasotraline in children with attention-deficit/hyperactivity disorder: A six-week, placebo-controlled, fixed-dose trial. J Child Adolesc Psychopharmacol . 2019;29(2):80–89. doi: 10.1089/cap.2018.0083. doi: [DOI] [PubMed] [Google Scholar]
  • 72.Wigal S, Tsai J, Bates JA et al. A randomized, placebo-controlled laboratory classroom study of the efficacy and safety of dasotraline in children with ADHD. J Atten Disord . 2020;26(10):1357–1368. doi: 10.1177/10870547211073477. doi: [DOI] [PubMed] [Google Scholar]
  • 73.Adler LA, Goldman R, Hopkins SC et al. Dasotraline in adults with attention deficit hyperactivity disorder: A placebo-controlled, fixed-dose trial. Int Clin Psychopharmacol . 2021;36(3):117–125. doi: 10.1097/YIC.0000000000000333. doi: [DOI] [PubMed] [Google Scholar]
  • 74.Sunovion. FDA issues a complete response letter for new drug application for dasotraline for the treatment of ADHD. 2018 August 31; https://news.sunovion.com/press-releases/press-releases-details/2018/FDA-Issues-a-Complete-Response-Letter-for-New-Drug-Application-for-Dasotraline-for-the-Treatment-of-ADHD/default.aspx#:~:text=Upon%20completion%20of%20their%20review,for%20the%20treatment%20of%20ADHD . [Google Scholar]
  • 75.Sunovion. Sunovion discontinues dasotraline program. 2020 May 13; https://news.sunovion.com/press-releases/press-releases-details/2020/Sunovion-Discontinues-Dasotraline-Program/default.aspx . [Google Scholar]
  • 76. CYMBALTA (duloxetine delayed-release capsules), for oral use [prescribing information]. Revised 8/2023. https://pi.lilly.com/us/cymbalta-pi.pdf. [Google Scholar]
  • 77.Bilodeau M, Simon T, Beauchamp MH et al. Duloxetine in adults with ADHD: A randomized, placebo-controlled pilot study. J Atten Disord . 2014;18(2):169–175. doi: 10.1177/1087054712443157. doi: [DOI] [PubMed] [Google Scholar]
  • 78.Dwoskin LP, Crooks PA. A novel mechanism of action and potential use for lobeline as a treatment for psychostimulant abuse. Biochem Pharmacol . 2002;63(2):89–98. doi: 10.1016/s0006-2952(01)00899-1. doi: [DOI] [PubMed] [Google Scholar]
  • 79.Zheng G, Horton DB, Deaciuc AG, Dwoskin LP, Crooks PA. Des-keto lobeline analogs with increased potency and selectivity at dopamine and serotonin transporters. Bioorg Med Chem Lett . 2006;16(19):5018–5021. doi: 10.1016/j.bmcl.2006.07.070. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Martin CA, Nuzzo PA, Ranseen JD et al. Lobeline effects on cognitive performance in adult ADHD. J Atten Disord . 2018;22(14):1361–1366. doi: 10.1177/1087054713497791. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. SANOREX® (mazindol) 1 and 2 mg tablets [product monograph]. Revised 7/2015. https://pdf.hres.ca/dpd_pm/00031211.PDF. [Google Scholar]
  • 82.Corser B, Stern T, Bogan R et al. 0585 A four-week randomized, double-blind, placebo-controlled, phase 2 study of mazindol ER in the treatment of narcolepsy. Sleep . 2023;46(Supplement_1):A257–A257. doi: 10.1093/sleep/zsad077.0585. doi: [DOI] [Google Scholar]
  • 83.Wigal TL, Newcorn JH, Handal N et al. A double-blind, placebo-controlled, phase II study to determine the efficacy, safety, tolerability and pharmacokinetics of a controlled release (CR) formulation of mazindol in adults with DSM-5 attention-deficit/hyperactivity disorder (ADHD) CNS Drugs . 2018;32(3):289–301. doi: 10.1007/s40263-018-0503-y. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.NLS Pharmaceutics news release. 2023 July 3; NLS Pharmaceutics to proceed with phase 3 clinical program (AMAZE) for mazindol ER for the treatment of narcolepsy following FDA review and IRB approval of the full study protocol. https://feeds.issuerdirect.com/news-release.html?newsid=5667709303839703 . [Google Scholar]
  • 85. PROVIGIL® (modafinil) tablets, for oral use [prescribing information]. Revised 1/2015. https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/020717s037s038lbl.pdf. [Google Scholar]
  • 86.Ishizuka T, Murotani T, Yamatodani A. Action of modafinil through histaminergic and orexinergic neurons. Vitam Horm . 2012;89:259–278. doi: 10.1016/B978-0-12-394623-2.00014-7. doi: [DOI] [PubMed] [Google Scholar]
  • 87.Ferraro L, Fuxe K, Tanganelli S, Tomasini MC, Rambert FA, Antonelli T. Differential enhancement of dialysate serotonin levels in distinct brain regions of the awake rat by modafinil: Possible relevance for wakefulness and depression. J Neurosci Res . 2002;68(1):107–112. doi: 10.1002/jnr.10196. doi: [DOI] [PubMed] [Google Scholar]
  • 88.Wang SM, Han C, Lee SJ et al. Modafinil for the treatment of attention-deficit/hyperactivity disorder: A meta-analysis. J Psychiatr Res . 2017;84:292–300. doi: 10.1016/j.jpsychires.2016.09.034. doi: [DOI] [PubMed] [Google Scholar]
  • 89.NPR. FDA committee rejects ADHD use for modafinil. 2006 March 24; https://www.npr.org/2006/03/24/5298885/fda-committee-rejects-adhd-use-for-modafinil . [Google Scholar]
  • 90.Ballon JS, Feifel D. A systematic review of modafinil: Potential clinical uses and mechanisms of action. J Clin Psychiatry . 2006;67(4):554–566. doi: 10.4088/jcp.v67n0406. doi: [DOI] [PubMed] [Google Scholar]
  • 91.Esposito M, Cocimano G, Ministrieri F et al. Smart drugs and neuroenhancement: What do we know? Front Biosci (Landmark Ed) . 2021;26(8):347–359. doi: 10.52586/4948. doi: [DOI] [PubMed] [Google Scholar]
  • 92.Loland CJ, Mereu M, Okunola OM et al. R-modafinil (armodafinil): A unique dopamine uptake inhibitor and potential medication for psychostimulant abuse. Biol Psychiatry . 2012;72(5):405–413. doi: 10.1016/j.biopsych.2012.03.022. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wilens TE, Klint T, Adler L et al. A randomized controlled trial of a novel mixed monoamine reuptake inhibitor in adults with ADHD. Behav Brain Funct . 2008;4:24. doi: 10.1186/1744-9081-4-24. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.NS 2359. Adis Inight. [June 11, 2024]; Accessed. https://adisinsight.springer.com/drugs/800010231 . [Google Scholar]
  • 95.Heal DJ, Smith SL, Findling RL. ADHD: current and future therapeutics. Curr Top Behav Neurosci . 2012;9:361–390. doi: 10.1007/7854_2011_125. doi: [DOI] [PubMed] [Google Scholar]
  • 96.Sepracor Inc. Annual report. 2008 December 31; https://www.sec.gov/Archives/edgar/data/877357/000104746909001949/a2190020z10-k.htm . [Google Scholar]
  • 97.Supplementary Financial Data for the Third Quarter of the Year Ending March 31, 2010 Dainippon Sumitomo Pharma Co., Ltd. https://www.sumitomo-pharma.com/ir/library/financial_results_summary/assets/pdf/ebr20100203_4.pdf . [Google Scholar]
  • 98.Sramek JJ, Hardy LW, Bieck P et al. Exploratory biomarker study of the triple reuptake inhibitor SEP-432 compared to the dual reuptake inhibitor duloxetine in healthy normal subjects. CNS Neurosci Ther . 2016;22(5):404–412. doi: 10.1111/cns.12513. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Supplementary Financial Data for the Second Quarter of the Year Ending March 31, 2013. [July 17, 2024]; Accessed. https://www.sumitomo-pharma.com/ir/library/financial_results_summary/assets/pdf/ebr20121031_4.pdf . [Google Scholar]
  • 100.Shire Pharmaceuticals Group PLC. 2002 December 31; Annual report. http://getfilings.com/o0000950162-03-000641.html . [Google Scholar]
  • 101.Heal DJ, Gosden J, Smith SL. New drugs to treat ADHD: Opportunities and challenges in research and development. Curr Top Behav Neurosci . 2022;57:79–126. doi: 10.1007/7854_2022_332. doi: [DOI] [PubMed] [Google Scholar]
  • 102.EFFEXOR XR® (venlafaxine extended-release) capsules [prescribing information]. Revised 1/2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/020699s107lbl.pdf . [Google Scholar]
  • 103.Ghanizadeh A, Freeman RD, Berk M. Efficacy and adverse effects of venlafaxine in children and adolescents with ADHD: A systematic review of non-controlled and controlled trials. Rev Recent Clin Trials . 2013;8(1):2–8. doi: 10.2174/1574887111308010002. doi: [DOI] [PubMed] [Google Scholar]
  • 104.Findling RL, Schwartz MA, Flannery DJ, Manos MJ. Venlafaxine in adults with attention-deficit/hyperactivity disorder: An open clinical trial. J Clin Psychiatry . 1996;57(5):184–189. [PubMed] [Google Scholar]
  • 105.Amiri S, Farhang S, Ghoreishizadeh MA, Malek A, Mohammadzadeh S. Double-blind controlled trial of venlafaxine for treatment of adults with attention deficit/hyperactivity disorder. Hum Psychopharmacol . 2012;27(1):76–81. doi: 10.1002/hup.1274. doi: [DOI] [PubMed] [Google Scholar]
  • 106.Ninan PT. Use of venlafaxine in other psychiatric disorders. Depress Anxiety . 2000;12(Suppl 1):90–4. doi: 10.1002/1520-6394(2000)12:1+&#x0003c;90::AID-DA13&#x0003e;3.0.CO;2-B. doi: [DOI] [PubMed] [Google Scholar]
  • 107.Weisler RH, Pandina GJ, Daly EJ, Cooper K, Gassmann-Mayer C Investigators ATTS. Randomized clinical study of a histamine H3 receptor antagonist for the treatment of adults with attention-deficit hyperactivity disorder. CNS Drugs . 2012;26(5):421–434. doi: 10.2165/11631990-000000000-00000. doi: [DOI] [PubMed] [Google Scholar]
  • 108.Herring WJ, Wilens TE, Adler LA et al. Randomized controlled study of the histamine H3 inverse agonist MK-0249 in adult attention-deficit/hyperactivity disorder. J Clin Psychiatry . 2012;73(7):e891–e898. doi: 10.4088/JCP.11m07178. doi: [DOI] [PubMed] [Google Scholar]
  • 109.Herring WJ, Liu K, Hutzelmann J et al. Alertness and psychomotor performance effects of the histamine-3 inverse agonist MK-0249 in obstructive sleep apnea patients on continuous positive airway pressure therapy with excessive daytime sleepiness: A randomized adaptive crossover study. Sleep Med . 2013;14(10):955–963. doi: 10.1016/j.sleep.2013.04.010. doi: [DOI] [PubMed] [Google Scholar]
  • 110.Egan M, Yaari R, Liu L et al. Pilot randomized controlled study of a histamine receptor inverse agonist in the symptomatic treatment of AD. Curr Alzheimer Res . 2012;9(4):481–490. doi: 10.2174/156720512800492530. doi: [DOI] [PubMed] [Google Scholar]
  • 111.Riley TB, Overton PG. Enhancing the efficacy of 5-HT uptake inhibitors in the treatment of attention deficit hyperactivity disorder. Med Hypotheses . 2019;133:109407. doi: 10.1016/j.mehy.2019.109407. doi: [DOI] [PubMed] [Google Scholar]
  • 112. ADDERALL® CII (dextroamphetamine saccharate, amphetamine aspartate, dextroamphetamine sulfate and amphetamine sulfate tablets) [prescribing information]. Revised 1/2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/011522s043lbl.pdf. [Google Scholar]
  • 113. EVEKEO® (amphetamine sulfate tablets, USP) [prescribing information]. Revised 4/2019. https://www.evekeo.com/pdfs/evekeo-pi.pdf. [Google Scholar]
  • 114. ZENZEDI® (dextroamphetamine sulfate tablets, USP) [prescribing information]. Revised 2/2017. https://zenzedi.com/docs/PIandMedicationGuide.pdf. [Google Scholar]
  • 115.Kuczenski R, Segal DS. Effects of methylphenidate on extracellular dopamine, serotonin, and norepinephrine: comparison with amphetamine. J Neurochem . 1997;68(5):2032–2037. doi: 10.1046/j.1471-4159.1997.68052032.x. doi: [DOI] [PubMed] [Google Scholar]
  • 116.Jones S, Kauer JA. Amphetamine depresses excitatory synaptic transmission via serotonin receptors in the ventral tegmental area. J Neurosci . 1999;19(22):9780–9787. doi: 10.1523/JNEUROSCI.19-22-09780.1999. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Berman SM, Kuczenski R, McCracken JT, London ED. Potential adverse effects of amphetamine treatment on brain and behavior: a review. Mol Psychiatry . 2009;14(2):123–142. doi: 10.1038/mp.2008.90. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Torres-Acosta N, O’Keefe JH, O’Keefe CL, Lavie CJ. Cardiovascular effects of ADHD therapies: JACC review topic of the week. J Am Coll Cardiol . 2020;76(7):858–866. doi: 10.1016/j.jacc.2020.05.081. doi: [DOI] [PubMed] [Google Scholar]
  • 119.Castells X, Blanco-Silvente L, Cunill R. Amphetamines for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev . 2018;8(8):CD007813. doi: 10.1002/14651858.CD007813.pub3. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. CONCERTA® (methylphenidate HCl) Extended-Release Tablets CII [prescribing information]. Revised 10/2023. https://www.janssenlabels.com/package-insert/product-monograph/prescribing-information/CONCERTA-pi.pdf. [Google Scholar]
  • 121. RITALIN® hydrochloride (methylphenidate hydrochloride USP) tablets and sustained-release tablets [prescribing information]. Revised 2013. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/010187s077lbl.pdf. [Google Scholar]
  • 122.Gatley SJ, Pan D, Chen R, Chaturvedi G, Ding YS. Affinities of methylphenidate derivatives for dopamine, norepinephrine and serotonin transporters. Life Sci . 1996;58(12):231–239. doi: 10.1016/0024-3205(96)00052-5. doi: [DOI] [PubMed] [Google Scholar]
  • 123.Markowitz JS, DeVane CL, Ramamoorthy S, Zhu HJ. The psychostimulant d-threo-(R,R)-methylphenidate binds as an agonist to the 5HT(1A) receptor. Pharmazie . 2009;64(2):123–125. [PubMed] [Google Scholar]
  • 124.Berger A. Ritalin may influence serotonin balance in hyperactive children. BMJ . 1999;318(7178):212B. doi: 10.1136/bmj.318.7178.212b. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Gainetdinov RR, Wetsel WC, Jones SR, Levin ED, Jaber M, Caron MG. Role of serotonin in the paradoxical calming effect of psychostimulants on hyperactivity. Science . 1999;283(5400):397–401. doi: 10.1126/science.283.5400.397. doi: [DOI] [PubMed] [Google Scholar]
  • 126.Molina-Carballo A, Naranjo-Gomez A, Uberos J et al. Methylphenidate effects on blood serotonin and melatonin levels may help to synchronise biological rhythms in children with ADHD. J Psychiatr Res . 2013;47(3):377–383. doi: 10.1016/j.jpsychires.2012.09.020. doi: [DOI] [PubMed] [Google Scholar]
  • 127.Ching C, Eslick GD, Poulton AS. Evaluation of methylphenidate safety and maximum-dose titration rationale in attention-deficit/hyperactivity disorder: A meta-analysis. JAMA Pediatr . 2019;173(7):630–639. doi: 10.1001/jamapediatrics.2019.0905. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Nemoda Z, Angyal N, Tarnok Z, Gadoros J, Sasvari-Szekely M. Carboxylesterase 1 gene polymorphism and methylphenidate response in ADHD. Neuropharmacology . 2009;57(7–8):731–733. doi: 10.1016/j.neuropharm.2009.08.014. doi: [DOI] [PubMed] [Google Scholar]
  • 129.Gajria K, Lu M, Sikirica V et al. Adherence, persistence, and medication discontinuation in patients with attention-deficit/hyperactivity disorder—a systematic literature review. Neuropsychiatr Dis Treat . 2014;10:1543–1569. doi: 10.2147/NDT.S65721. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. STRATTERA® (atomoxetine) capsules, for oral use [prescribing information]. Revised 1/2022. https://pi.lilly.com/us/strattera-pi.pdf. [Google Scholar]
  • 131.Ding YS, Naganawa M, Gallezot JD et al. Clinical doses of atomoxetine significantly occupy both norepinephrine and serotonin transports: Implications on treatment of depression and ADHD. Neuroimage . 2014;86:164–171. doi: 10.1016/j.neuroimage.2013.08.001. doi: [DOI] [PubMed] [Google Scholar]
  • 132.Michelson D, Adler L, Spencer T et al. Atomoxetine in adults with ADHD: Two randomized, placebo-controlled studies. Biol Psychiatry . 2003;53(2):112–120. doi: 10.1016/s0006-3223(02)01671-2. doi: [DOI] [PubMed] [Google Scholar]
  • 133.Purper-Ouakil D, Fourneret P, Wohl M, Reneric JP. Atomoxetine: A new treatment for attention deficit/hyperactivity disorder (ADHD) in children and adolescents. Encephale . 2005;31(3):337–348. doi: 10.1016/s0013-7006(05)82399-1. L’atomoxetine: un nouveau traitement du trouble deficitaire de l’attention/hyperactivite de l’enfant et de l’adolescent. doi: [DOI] [PubMed] [Google Scholar]
  • 134. QELBREE® (viloxazine extended-release capsules), for oral use [prescribing information]. Revised 4/2022. https://www.supernus.com/sites/default/files/Qelbree-Prescribing-Info.pdf. [Google Scholar]
  • 135.Yu C, Garcia-Olivares J, Candler S, Schwabe S, Maletic V. New insights into the mechanism of action of viloxazine: Serotonin and norepinephrine modulating properties. J Exp Pharmacol . 2020;12:285–300. doi: 10.2147/JEP.S256586. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Bubar MJ, Stutz SJ, Cunningham KA. 5-HT2C receptors localize to dopamine and GABA neurons in the rat mesoaccumbens pathway. PLoS One . 2011;6(6):e20508. doi: 10.1371/journal.pone.0020508. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.De Deurwaerdere P, Chagraoui A, Di Giovanni G. Serotonin/dopamine interaction: Electrophysiological and neurochemical evidence. Prog Brain Res . 2021;261:161–264. doi: 10.1016/bs.pbr.2021.02.001. doi: [DOI] [PubMed] [Google Scholar]
  • 138.Garcia-Olivares J, Yegla B, Earnest J, Yu C, Rubin J. P175. Unraveling the mechanism of action of viloxazine ER in ADHD. Neuropsychopharmacol . 2023;48(Suppl 1):166. Poster presented at the American College of Neuropsychopharmacology 62nd Annual Meeting (Dec 03–06, 2023; Tampa, Florida) [Google Scholar]
  • 139.Robinson ES, Dalley JW, Theobald DE et al. Opposing roles for 5-HT2A and 5-HT2C receptors in the nucleus accumbens on inhibitory response control in the 5-choice serial reaction time task. Neuropsychopharmacology . 2008;33(10):2398–2406. doi: 10.1038/sj.npp.1301636. doi: [DOI] [PubMed] [Google Scholar]
  • 140.Pisansky MT, Lefevre EM, Retzlaff CL, Trieu BH, Leipold DW, Rothwell PE. Nucleus accumbens fast-spiking interneurons constrain impulsive action. Biol Psychiatry . 2019;86(11):836–847. doi: 10.1016/j.biopsych.2019.07.002. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. VALDOXAN (agomelatine) 25 mg film-coated tablets [summary of product characteristics]. Revised 12/2018. https://www.ema.europa.eu/en/documents/product-information/valdoxan-epar-product-information_en.pdf. [Google Scholar]
  • 142.Millan MJ, Gobert A, Lejeune F et al. The novel melatonin agonist agomelatine (S20098) is an antagonist at 5-hydroxytryptamine2C receptors, blockade of which enhances the activity of frontocortical dopaminergic and adrenergic pathways. J Pharmacol Exp Ther . 2003;306(3):954–964. doi: 10.1124/jpet.103.051797. doi: [DOI] [PubMed] [Google Scholar]
  • 143.Savino R, Polito AN, Marsala G et al. Agomelatine: A potential multitarget compound for neurodevelopmental disorders. Brain Sci . 2023;13(5):734. doi: 10.3390/brainsci13050734. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Niederhofer H. Agomelatine treatment with adolescents with ADHD. J Atten Disord . 2012;16(6):530–2. doi: 10.1177/1087054711423631. doi: [DOI] [PubMed] [Google Scholar]
  • 145.Salardini E, Zeinoddini A, Kohi A et al. Agomelatine as a treatment for attention-deficit/hyperactivity disorder in children and adolescents: A double-blind, randomized clinical trial. J Child Adolesc Psychopharmacol . 2016;26(6):513–519. doi: 10.1089/cap.2016.0024. doi: [DOI] [PubMed] [Google Scholar]
  • 146. NORPRAMIN® (desipramine hydrochloride tablets) [prescribing information]. Revised 6/2014. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/014399s069lbl.pdf. [Google Scholar]
  • 147.Chung B, Suzuki AR, McGough JJ. New drugs for treatment of attention-deficit/hyperactivity disorder. Expert Opin Emerg Drugs . 2002;7(2):269–276. doi: 10.1517/14728214.7.2.269. doi: [DOI] [PubMed] [Google Scholar]
  • 148.Maan JS, Rosani A, Saadabadi A. Desipramine. StatPearls . 2024 [PubMed] [Google Scholar]
  • 149.Cowen PJ, Geaney DP, Schachter M, Green AR, Elliott JM. Desipramine treatment in normal subjects. Effects on neuroendocrine responses to tryptophan and on platelet serotonin (5-HT)-related receptors. Arch Gen Psychiatry . 1986;43(1):61–67. doi: 10.1001/archpsyc.1986.01800010063008. doi: [DOI] [PubMed] [Google Scholar]
  • 150.Ghanizadeh A. A systematic review of the efficacy and safety of desipramine for treating ADHD. Curr Drug Saf . 2013;8(3):169–174. doi: 10.2174/15748863113089990029. doi: [DOI] [PubMed] [Google Scholar]
  • 151. Imipramine hydrochloride tablet [prescribing information]. Revised 10/2012. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/040903lbl.pdf. [Google Scholar]
  • 152.Budur K, Mathews M, Adetunji B, Mathews M, Mahmud J. Non-stimulant treatment for attention deficit hyperactivity disorder. Psychiatry (Edgmont) . 2005;2(7):44–48. [PMC free article] [PubMed] [Google Scholar]
  • 153.Fayez R, Gupta V. Imipramine. StatPearls . 2024 https://www.ncbi.nlm.nih.gov/books/NBK557656/ [PubMed] [Google Scholar]
  • 154.Bijak M, Tokarski K, Czyrak A, Mackowiak M, Wedzony K. Imipramine increases the 5-HT1A receptor-mediated inhibition of hippocampal neurons without changing the 5-HT1A receptor binding. Eur J Pharmacol . 1996;305(1–3):79–85. doi: 10.1016/0014-2999(96)00151-3. doi: [DOI] [PubMed] [Google Scholar]
  • 155. VRAYLAR® (cariprazine) capsules, for oral use [prescribing information]. Revised 2/2024. https://www.rxabbvie.com/pdf/vraylar_pi.pdf. [Google Scholar]
  • 156.Otsuka. Press release. Otsuka pharmaceutical announces positive topline results from two pivotal phase 3 trials of centanafadine as a treatment for adolescents and children with attention-deficit/hyperactivity disorder (ADHD) 2023 October 27; https://www.otsuka-us.com/news/otsuka-pharmaceutical-announces-positive-topline-results-two-pivotal-phase-3-trials#:~:text=Phase%203%20Program-,Centanafadine%20is%20a%20first%2Din%2Dclass%20norepinephrine%2C%20dopamine%2C,children%2C%20adolescents%2C%20and%20adults . [Google Scholar]
  • 157.Wigal SB, Wigal T, Hobart M et al. Safety and efficacy of centanafadine sustained-release in adults with attention-deficit hyperactivity disorder: Results of phase 2 studies. Neuropsychiatr Dis Treat . 2020;16:1411–1426. doi: 10.2147/NDT.S242084. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Adler LA, Adams J, Madera-McDonough J et al. Efficacy, safety, and tolerability of centanafadine sustained-release tablets in adults with attention-deficit/hyperactivity disorder: Results of 2 phase 3, randomized, double-blind, multicenter, placebo-controlled trials. J Clin Psychopharmacol . 2022;42(5):429–439. doi: 10.1097/JCP.0000000000001575. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. SUNOSI (solriamfetol) tablets, for oral use [prescribing information]. Revised 3/2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/211230s000lbl.pdf. [Google Scholar]
  • 160.Gursahani H, Jolas T, Martin M et al. Preclinical pharmacology of solriamfetol: Potential mechanisms for wake promotion (P4-13.005) Neurology . 2023;100(suppl 2) doi: 10.1212/WNL.0000000000204178. doi: [DOI] [Google Scholar]
  • 161.Revel FG, Moreau JL, Gainetdinov RR et al. TAAR1 activation modulates monoaminergic neurotransmission, preventing hyperdopaminergic and hypoglutamatergic activity. Proc Natl Acad Sci U S A . 2011;108(20):8485–8490. doi: 10.1073/pnas.1103029108. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Underhill S, Amara S. MDMA and TAAR1-mediated RhoA activation in serotonin neurons. FASEB J . 2020;34(S1):1–1. doi: 10.1096/fasebj.2020.34.s1.05856. doi: [DOI] [Google Scholar]
  • 163.Zhukov IS, Karpova IV, Krotova NA et al. Enhanced aggression, reduced self-grooming behavior and altered 5-HT regulation in the frontal cortex in mice lacking trace amine-associated receptor 1 (TAAR1) Int J Mol Sci . 2022;23(22):14066. doi: 10.3390/ijms232214066. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Surman CBH, Walsh DM, Horick N, DiSalvo M, Vater CH, Kaufman D. Solriamfetol for attention-deficit/hyperactivity disorder in adults: A double-blind placebo-controlled pilot study. J Clin Psychiatry . 2023;84(6):23m14934. doi: 10.4088/JCP.23m14934. doi: [DOI] [PubMed] [Google Scholar]
  • 164.Ayano G, Tsegay L, Gizachew Y et al. Prevalence of attention deficit hyperactivity disorder in adults: Umbrella review of evidence generated across the globe. Psychiatry Res . 2023;328:115449. doi: 10.1016/j.psychres.2023.115449. doi: [DOI] [PubMed] [Google Scholar]
  • 166.Cutler AJ, Mattingly GW, Jain R, O’Neal W. Current and future nonstimulants in the treatment of pediatric ADHD: Monoamine reuptake inhibitors, receptor modulators, and multimodal agents. CNS Spectr . 2022;27(2):199–207. doi: 10.1017/S1092852920001984. doi: [DOI] [PubMed] [Google Scholar]

References for Table 1. Interventions for ADHD with a Serotonergic Mechanism of Action That Failed in the ADHD Indication

  • 1.Rammes G, Rupprecht R, Ferrari U, Zieglgansberger W, Parsons CG. The N-methyl-D-aspartate receptor channel blockers memantine, MRZ 2/579 and other amino-alkyl-cyclohexanes antagonise 5-HT(3) receptor currents in cultured HEK-293 and N1E-115 cell systems in a non-competitive manner. Neurosci Lett . 2001;306(1–2):81–84. doi: 10.1016/s0304-3940(01)01872-9. doi: [DOI] [PubMed] [Google Scholar]
  • 2.Lemmer B. Effects of amantadine and amphetamine on serotonin uptake and release by human blood platelets. Eur J Pharmacol . 1973;21(2):183–188. doi: 10.1016/0014-2999(73)90224-0. doi: [DOI] [PubMed] [Google Scholar]
  • 3.Wesemann W, Dette-Wildenhahn G, Fellehner H. In vitro studies on the possible effects of 1-aminoadamantanes on the serotonergic system in M. Parkinson. J Neural Transm . 1979;44(4):263–285. doi: 10.1007/bf01250322. doi: [DOI] [PubMed] [Google Scholar]
  • 4. GOCOVRITM (amantadine) extended release capsules, for oral use [prescribing information]. Revised 8/2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/208944lbl.pdf. [Google Scholar]
  • 5.Weisler RH, Pandina GJ, Daly EJ, Cooper K, Gassmann-Mayer C. Investigators ATTS. Randomized clinical study of a histamine H3 receptor antagonist for the treatment of adults with attention-deficit hyperactivity disorder. CNS Drugs . 2012;26(5):421–434. doi: 10.2165/11631990-000000000-00000. doi: [DOI] [PubMed] [Google Scholar]
  • 6. REXULTI® (brexpiprazole) tablets, for oral use [prescribing information]. Revised 5/2023. https://www.otsuka-us.com/sites/g/files/qhldwo8366/files/media/static/Rexulti-PI.pdf. [Google Scholar]
  • 7.Maeda K, Sugino H, Akazawa H et al. Brexpiprazole I: in vitro and in vivo characterization of a novel serotonin-dopamine activity modulator. J Pharmacol Exp Ther . 2014;350(3):589–604. doi: 10.1124/jpet.114.213793. doi: [DOI] [PubMed] [Google Scholar]
  • 8.2024 Psychoactive Drug Screening Program PDSP Ki Database. https://pdsp.unc.edu/databases/kidb.php . [Google Scholar]
  • 9.Schreiber R, Campbell U, Quinton MS, Hardy LW, Fang QK, Lew R. In vitro and in vivo pharmacological characterization of dasotraline, a dual dopamine and norepinephrine transporter inhibitor in vivo. Biomed Pharmacother . 2022;153:113359. doi: 10.1016/j.biopha.2022.113359. doi: [DOI] [PubMed] [Google Scholar]
  • 10. [June 7, 2024]; FDA rejects dasotraline NDA for ADHD, calling for more data. ThePharmaLetter. Accessed. https://www.thepharmaletter.com/article/fda-rejects-dasotraline-nda-for-adhd-calling-for-more-data . [Google Scholar]
  • 11.2020 May 13; Sunovion. Sunovion discontinues dasotraline program. https://news.sunovion.com/press-releases/press-releases-details/2020/Sunovion-Discontinues-Dasotraline-Program/default.aspx . [Google Scholar]
  • 12.Han M, Song C, Jeong N, Hahn HG. Exploration of 3-Aminoazetidines as Triple Reuptake Inhibitors by Bioisosteric Modification of 3-α-Oxyazetidine. ACS Med Chem Lett . 2014;5(9):999–1004. doi: 10.1021/ml500187a. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Fish PV, Fray MJ, Stobie A, Wakenhut F, Whitlock GA. N-Benzyl-N-(tetrahydro-2H-pyran-4-yl)pyrrolidin-3-amines as selective dual serotonin/noradrenaline reuptake inhibitors. Bioorg Med Chem Lett . 2007;17(7):2022–2025. doi: 10.1016/j.bmcl.2007.01.020. doi: [DOI] [PubMed] [Google Scholar]
  • 14.Sabatucci JP, Mahaney PE, Leiter J et al. Heterocyclic cycloalkanol ethylamines as norepinephrine reuptake inhibitors. Bioorg Med Chem Lett . 2010;20(9):2809–2812. doi: 10.1016/j.bmcl.2010.03.059. doi: [DOI] [PubMed] [Google Scholar]
  • 15.McComas CC, Vu AT, Mahaney PE et al. Synthesis and activity of 1-(3-amino-1-phenylpropyl)indolin-2-ones: a new class of selective norepinephrine reuptake inhibitors. Bioorg Med Chem Lett . 2008;18(18):4929–4931. doi: 10.1016/j.bmcl.2008.08.060. doi: [DOI] [PubMed] [Google Scholar]
  • 16.Vu AT, Cohn ST, Terefenko EA et al. 3-(Arylamino)-3-phenylpropan-2-olamines as a new series of dual norepinephrine and serotonin reuptake inhibitors. Bioorg Med Chem Lett . 2009;19(9):2464–2467. doi: 10.1016/j.bmcl.2009.03.054. doi: [DOI] [PubMed] [Google Scholar]
  • 17.Dyck B, Tamiya J, Jovic F et al. Characterization of thien-2-yl 1S,2R-milnacipran analogues as potent norepinephrine/serotonin transporter inhibitors for the treatment of neuropathic pain. J Med Chem . 2008;51(22):7265–7272. doi: 10.1021/jm8009537. doi: [DOI] [PubMed] [Google Scholar]
  • 18.Wong DT, Bymaster FP, Mayle DA, Reid LR, Krushinski JH, Robertson DW. LY248686, a new inhibitor of serotonin and norepinephrine uptake. Neuropsychopharmacology . 1993;8(1):23–33. doi: 10.1038/npp.1993.4. doi: [DOI] [PubMed] [Google Scholar]
  • 19.Lucas MC, Carter DS, Cai HY et al. Novel, achiral aminoheterocycles as selective monoamine reuptake inhibitors. Bioorg Med Chem Lett . 2009;19(16):4630–4633. doi: 10.1016/j.bmcl.2009.06.076. doi: [DOI] [PubMed] [Google Scholar]
  • 20.Vaishnavi SN, Nemeroff CB, Plott SJ, Rao SG, Kranzler J, Owens MJ. Milnacipran: A comparative analysis of human monoamine uptake and transporter binding affinity. Biol Psychiatry . 2004;55(3):320–322. doi: 10.1016/j.biopsych.2003.07.006. doi: [DOI] [PubMed] [Google Scholar]
  • 21.Bymaster FP, Dreshfield-Ahmad LJ, Threlkeld PG et al. Comparative affinity of duloxetine and venlafaxine for serotonin and norepinephrine transporters in vitro and in vivo, human serotonin receptor subtypes, and other neuronal receptors. Neuropsychopharmacology . 2001;25(6):871–880. doi: 10.1016/s0893-133x(01)00298-6. doi: [DOI] [PubMed] [Google Scholar]
  • 22.Kuo F, Gillespie TA, Kulanthaivel P et al. Synthesis and biological activity of some known and putative duloxetine metabolites. Bioorg Med Chem Lett . 2004;14(13):3481–3486. doi: 10.1016/j.bmcl.2004.04.066. doi: [DOI] [PubMed] [Google Scholar]
  • 23.Fish PV, Deur C, Gan X et al. Design and synthesis of morpholine derivatives. SAR for dual serotonin & noradrenaline reuptake inhibition. Bioorg Med Chem Lett . 2008;18(8):2562–2566. doi: 10.1016/j.bmcl.2008.03.050. doi: [DOI] [PubMed] [Google Scholar]
  • 24.Auclair AL, Martel JC, Assié MB. Levomilnacipran (F2695), a norepinephrine-preferring SNRI: profile in vitro and in models of depression and anxiety. Neuropharmacology . 2013;70:338–347. doi: 10.1016/j.neuropharm.2013.02.024. doi: [DOI] [PubMed] [Google Scholar]
  • 25.Béïque JC, Lavoie N, de Montigny C, Debonnel G. Affinities of venlafaxine and various reuptake inhibitors for the serotonin and norepinephrine transporters. Eur J Pharmacol . 1998;349(1):129–132. doi: 10.1016/s0014-2999(98)00241-6. doi: [DOI] [PubMed] [Google Scholar]
  • 26.Manning JR, Sexton T, Childers SR, Davies HM. 1-Naphthyl and 4-indolyl arylalkylamines as selective monoamine reuptake inhibitors. Bioorg Med Chem Lett . 2009;19(1):58–61. doi: 10.1016/j.bmcl.2008.11.022. doi: [DOI] [PubMed] [Google Scholar]
  • 27. CYMBALTA (duloxetine delayed-release capsules), for oral use [prescribing information]. Revised 8/2023. https://pi.lilly.com/us/cymbalta-pi.pdf. [Google Scholar]
  • 28.Miller DK, Crooks PA, Zheng G, Grinevich VP, Norrholm SD, Dwoskin LP. Lobeline analogs with enhanced affinity and selectivity for plasmalemma and vesicular monoamine transporters. J Pharmacol Exp Ther . 2004;310(3):1035–1045. doi: 10.1124/jpet.104.068098. doi: [DOI] [PubMed] [Google Scholar]
  • 29.Zheng G, Horton DB, Deaciuc AG, Dwoskin LP, Crooks PA. Des-keto lobeline analogs with increased potency and selectivity at dopamine and serotonin transporters. Bioorg Med Chem Lett . 2006;16(19):5018–5021. doi: 10.1016/j.bmcl.2006.07.070. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hojahmat M, Horton DB, Norrholm SD et al. Lobeline esters as novel ligands for neuronal nicotinic acetylcholine receptors and neurotransmitter transporters. Bioorg Med Chem . 2010;18(2):640–649. doi: 10.1016/j.bmc.2009.12.002. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Provencher BA, Eshleman AJ, Johnson RA et al. Synthesis and Discovery of Arylpiperidinylquinazolines: New Inhibitors of the Vesicular Monoamine Transporter. J Med Chem . 2018;61(20):9121–9131. doi: 10.1021/acs.jmedchem.8b00542. doi: [DOI] [PubMed] [Google Scholar]
  • 32.Houlihan WJ, Boja JW, Parrino VA, Kopajtic TA, Kuhar MJ. Halogenated mazindol analogs as potential inhibitors of the cocaine binding site at the dopamine transporter. J Med Chem . 1996;39(25):4935–4941. doi: 10.1021/jm960288w. doi: [DOI] [PubMed] [Google Scholar]
  • 33.Barker EL, Perlman MA, Adkins EM et al. High affinity recognition of serotonin transporter antagonists defined by species-scanning mutagenesis. An aromatic residue in transmembrane domain I dictates species-selective recognition of citalopram and mazindol. J Biol Chem . 1998;273(31):19459–19468. doi: 10.1074/jbc.273.31.19459. doi: [DOI] [PubMed] [Google Scholar]
  • 34.Kung MP, Essman WD, Frederick D et al. IPT: a novel iodinated ligand for the CNS dopamine transporter. Synapse . 1995;20(4):316–324. doi: 10.1002/syn.890200405. doi: [DOI] [PubMed] [Google Scholar]
  • 35.Rothman RB, Baumann MH, Dersch CM et al. Amphetamine-type central nervous system stimulants release norepinephrine more potently than they release dopamine and serotonin. Synapse . 2001;39(1):32–41. doi: 10.1002/1098-2396(20010101)39:1&#x0003c;32::Aid-syn5&#x0003e;3.0.Co;2-3. doi: [DOI] [PubMed] [Google Scholar]
  • 36.Tatsumi M, Groshan K, Blakely RD, Richelson E. Pharmacological profile of antidepressants and related compounds at human monoamine transporters. Eur J Pharmacol . 1997;340(2–3):249–258. doi: 10.1016/s0014-2999(97)01393-9. doi: [DOI] [PubMed] [Google Scholar]
  • 37. SANOREX® (mazindol) 1 and 2 mg tablets [product monograph]. Revised 7/2015. https://pdf.hres.ca/dpd_pm/00031211.PDF. [Google Scholar]
  • 38. [June 7, 2024]; NLS Pharmaceutics to Proceed with Phase 3 Clinical Program (AMAZE) for Mazindol ER for the Treatment of Narcolepsy Following FDA Review and IRB Approval of the Full Study Protocol. Accessed. https://www.sec.gov/Archives/edgar/data/1783036/000121390023053971/ea181301ex99-3_nlspharma.htm . [Google Scholar]
  • 39. NAMENDA® (memantine hydrochloride) tablets, for oral use [prescribing information]. Revised 11/2018. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/021487s025lbl.pdf. [Google Scholar]
  • 40.Herring WJ, Wilens TE, Adler LA et al. Randomized controlled study of the histamine H3 inverse agonist MK-0249 in adult attention-deficit/hyperactivity disorder. J Clin Psychiatry . 2012;73(7):e891–e898. doi: 10.4088/JCP.11m07178. doi: [DOI] [PubMed] [Google Scholar]
  • 41.Kalaba P, Ilić M, Aher NY et al. Structure-activity relationships of novel thiazole-based modafinil analogues acting at monoamine transporters. J Med Chem . 2020;63(1):391–417. doi: 10.1021/acs.jmedchem.9b01938. doi: [DOI] [PubMed] [Google Scholar]
  • 42.Madras BK, Xie Z, Lin Z et al. Modafinil occupies dopamine and norepinephrine transporters in vivo and modulates the transporters and trace amine activity in vitro. J Pharmacol Exp Ther . 2006;319(2):561–569. doi: 10.1124/jpet.106.106583. doi: [DOI] [PubMed] [Google Scholar]
  • 43.Louvet P, Schweizer D, Gourdel ME et al. Wake-promoting agents: search for next generation modafinil: part IV. Eur J Med Chem . 2012;54:949–951. doi: 10.1016/j.ejmech.2012.05.038. doi: [DOI] [PubMed] [Google Scholar]
  • 44.Slack RD, Ku TC, Cao J et al. Structure-activity relationships for a series of (Bis(4-fluorophenyl)methyl)sulfinyl alkyl alicyclic amines at the dopamine transporter: Functionalizing the terminal nitrogen affects affinity, selectivity, and metabolic stability. J Med Chem . 2020;63(5):2343–2357. doi: 10.1021/acs.jmedchem.9b01188. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. PROVIGIL® (modafinil) tablets, for oral use [prescribing information]. Revised 1/2015. https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/020717s037s038lbl.pdf. [Google Scholar]
  • 46.FDA committee rejects ADHD use for modafinil. 2006 March 24; NPR. https://www.npr.org/2006/03/24/5298885/fda-committee-rejects-adhd-use-for-modafinil . [Google Scholar]
  • 47.Yu C, Gopalakrishnan G. In vitro pharmacological characterization of SPN-810M (molindone) J Exp Pharmacol . 2018;10:65–73. doi: 10.2147/JEP.S180777. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kroeze WK, Hufeisen SJ, Popadak BA et al. H1-histamine receptor affinity predicts short-term weight gain for typical and atypical antipsychotic drugs. Neuropsychopharmacology . 2003;28(3):519–526. doi: 10.1038/sj.npp.1300027. doi: [DOI] [PubMed] [Google Scholar]
  • 49.Roth BL, Craigo SC, Choudhary MS et al. Binding of typical and atypical antipsychotic agents to 5-hydroxytryptamine-6 and 5-hydroxytryptamine-7 receptors. J Pharmacol Exp Ther . 1994;268(3):1403–1410. [PubMed] [Google Scholar]
  • 50.Seeman P, Tallerico T. Antipsychotic drugs which elicit little or no parkinsonism bind more loosely than dopamine to brain D2 receptors, yet occupy high levels of these receptors. Mol Psychiatry . 1998;3(2):123–134. doi: 10.1038/sj.mp.4000336. doi: [DOI] [PubMed] [Google Scholar]
  • 51.Glennon RA. Higher-end serotonin receptors: 5-HT(5), 5-HT(6), and 5-HT(7) J Med Chem . 2003;46(14):2795–2812. doi: 10.1021/jm030030n. doi: [DOI] [PubMed] [Google Scholar]
  • 52. [June 7, 2024]; Supernus Pharma’s SPN-810 trial did not meet primary endpoint. Seeking Alpa. Accessed. https://seekingalpha.com/news/3545617-supernus-pharmas-spnminus-810-trial-not-meet-primary-endpoint . [Google Scholar]
  • 53.Subbaiah MAM. Triple reuptake inhibitors as potential therapeutics for depression and other disorders: Design paradigm and developmental challenges. J Med Chem . 2018;61(6):2133–2165. doi: 10.1021/acs.jmedchem.6b01827. doi: [DOI] [PubMed] [Google Scholar]
  • 54.Tu G, Fu T, Yang F et al. Understanding the polypharmacological profiles of triple reuptake inhibitors by molecular simulation. ACS Chem Neurosci . 2021;12(11):2013–2026. doi: 10.1021/acschemneuro.1c00127. doi: [DOI] [PubMed] [Google Scholar]
  • 55. [June 7, 2024]; NS 2359. AdisInsight. Accessed. https://adisinsight.springer.com/drugs/800010231 . [Google Scholar]
  • 56.Huot P, Fox SH, Brotchie JM. Monoamine reuptake inhibitors in Parkinson’s disease. Parkinsons Dis . 2015;2015:609428. doi: 10.1155/2015/609428. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Aggarwal S, Mortensen OV. Discovery and development of monoamine transporter ligands. Adv Neurobiol . 2023;30:101–129. doi: 10.1007/978-3-031-21054-9_4. doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Franz AK, Wilson SO. Organosilicon molecules with medicinal applications. J Med Chem . 2013;56(2):388–405. doi: 10.1021/jm3010114. doi: [DOI] [PubMed] [Google Scholar]
  • 59. EFFEXOR XR® (venlafaxine extended-release) capsules [prescribing information]. Revised 1/2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/020699s107lbl.pdf. [Google Scholar]
  • 60.2023 PRISTIQ® (desvenlafaxine) Prescribing Information. Pfizer. https://labeling.pfizer.com/showlabeling.aspx?id=497 . [Google Scholar]
  • 61. TRINTELLIX (vortioxetine) tablets, for oral use [prescribing information]. Revised 8/2023. https://content.takeda.com/?contenttype=PI&product=TRI&language=ENG&country=GBL&documentnumber=1. [Google Scholar]
  • 62.Bang-Andersen B, Ruhland T, Jørgensen M et al. Discovery of 1-[2-(2,4-dimethylphenylsulfanyl)phenyl]piperazine (Lu AA21004): a novel multimodal compound for the treatment of major depressive disorder. J Med Chem . 2011 May 12;54(9):3206–3221. doi: 10.1021/jm101459g. doi: [DOI] [PubMed] [Google Scholar]
  • 63.Gu ZS, Xiao Y, Zhang QW, Li JQ. Synthesis and antidepressant activity of a series of arylalkanol and aralkyl piperazine derivatives targeting SSRI/5-HT(1A)/5-HT(7) Bioorg Med Chem Lett . 2017;27(24):5420–5423. doi: 10.1016/j.bmcl.2017.11.007. doi: [DOI] [PubMed] [Google Scholar]
  • 64.Biederman J, Lindsten A, Sluth LB et al. Vortioxetine for attention deficit hyperactivity disorder in adults: A randomized, double-blind, placebo-controlled, proof-of-concept study. J Psychopharmacol . 2019;33(4):511–521. doi: 10.1177/0269881119832538. doi: [DOI] [PubMed] [Google Scholar]

Articles from Psychopharmacology Bulletin are provided here courtesy of MedWorks Media Inc.

RESOURCES