Next Article in Journal
Combination of Experimental and Bioinformatic Approaches for Identification of Immunologically Relevant Protein–Peptide Interactions
Next Article in Special Issue
The Constitutive Activity of Spinal 5-HT6 Receptors Contributes to Diabetic Neuropathic Pain in Rats
Previous Article in Journal
Knotify+: Toward the Prediction of RNA H-Type Pseudoknots, Including Bulges and Internal Loops
Previous Article in Special Issue
1-(Arylsulfonyl-isoindol-2-yl)piperazines as 5-HT6R Antagonists: Mechanochemical Synthesis, In Vitro Pharmacological Properties and Glioprotective Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Progress in Investigational Agents Targeting Serotonin-6 Receptors for the Treatment of Brain Disorders

1
Suven Life Sciences Limited, Serene Chambers, Road-5, Avenue-7, Banjara Hills, Hyderabad 500034, Telangana, India
2
Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, NV 89154, USA
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(2), 309; https://doi.org/10.3390/biom13020309
Submission received: 28 December 2022 / Revised: 3 February 2023 / Accepted: 4 February 2023 / Published: 7 February 2023
(This article belongs to the Special Issue Recent Advances on 5-HT6 Receptors)

Abstract

:
Serotonin (5-HT) plays an important role in the regulation of several basic functions of the central and peripheral nervous system. Among the 5-HT receptors, serotonin-6 (5-HT6) receptor has been an area of substantial research. 5-HT6 receptor is a G-protein-coupled receptor mediating its effects through diverse signaling pathways. Exceptional features of the receptors fueling drug discovery efforts include unique localization and specific distribution in the brain regions having a role in learning, memory, mood, and behavior, and the affinity of several clinically used psychotropic agents. Although non-clinical data suggest that both agonist and antagonist may have similar behavioral effects, most of the agents that entered clinical evaluation were antagonists. Schizophrenia was the initial target; more recently, cognitive deficits associated with Alzheimer’s disease (AD) or other neurological disorders has been the target for clinically evaluated 5-HT6 receptor antagonists. Several 5-HT6 receptor antagonists (idalopirdine, intepirdine and latrepirdine) showed efficacy in alleviating cognitive deficits associated with AD in the proof-of-concept clinical studies; however, the outcomes of the subsequent phase 3 studies were largely disappointing. The observations from both non-clinical and clinical studies suggest that 5-HT6 receptor antagonists may have a role in the management of neuropsychiatric symptoms in dementia. Masupirdine, a selective 5-HT6 receptor antagonist, reduced agitation/aggression-like behaviors in animal models, and a post hoc analysis of a phase 2 trial suggested potential beneficial effects on agitation/aggression and psychosis in AD. This agent will be assessed in additional trials, and the outcome of the trials will inform the use of 5-HT6 receptor antagonists in the treatment of agitation in dementia of the Alzheimer’s type.

1. Introduction

Serotonin-5-hydroxytryptamine (5-HT) or enteramine was discovered in the 1930s [1], and its presence in the brain was identified in the year 1953 [2]. Since then, advancements have been made in characterizing 5-HT and its receptors. 5-HT functions as an inhibitory monoamine neurotransmitter, a hormone, and a mitogen that plays an important role in the regulation of several basic functions of the central and peripheral nervous system [3]. The 5-HT transporter regulates the availability of 5-HT in the synapse. 5-HT mediates its functions through seven distinct families of receptors (5-HT1-7), which comprise 14 distinct receptor subtypes. Most of the 5-HT receptor types are G-protein coupled receptors (GPCRs), except for the 5-HT3 receptor, which is a ligand-gated cation channel. GPCRs can modulate diverse downstream signaling pathways, and this may partly explain the critical role of serotonin in controlling/modulating multiple physiological functions [4,5].
Only a fraction (~5%) of the total amount of 5-HT is produced in the brain [3,6], but 5-HT and 5-HT receptors play an important role in the regulation of many brain functions. In addition, the 5-HT system also interacts with other neurotransmitter systems in the brain [7]. Dysregulation of the 5-HT and/or 5-HT receptors has been implicated in the pathogenesis of several disorders including, but not limited to depression, anxiety, Alzheimer’s disease (AD), schizophrenia, Parkinson’s disease, and sleep disorders [8,9,10]. The majority of drugs (~40%) approved for use in humans for the treatment of various disorders target the 5-HT system [11]. The distribution of 5-HT receptors in the brain and diverse signaling pathways involved make the 5-HT system an important potential target for therapeutic interventions. The development and characterization of gene knock-out animals; expanding knowledge of receptor expression levels, specific localization, distribution and function; and progress in receptor subtype-specific agents comprise significant advancements in the area of 5-HT research. Considering the widespread distribution and functions of the serotonergic system, targeting the 5-HT receptors with specific characteristics and localization may define a drug development pathway. One of the 5-HT receptors—serotonin-6 (5-HT6) receptor—has been an area of substantial research.
The 5-HT6 receptor was first cloned and characterized from rat striatum by Monsma and colleagues in 1993 [12]. The human 5-HT6 receptor was cloned and characterized in 1996 by Kohen and colleagues [13]. Detailed review of the discovery and characterization of the 5-HT6 receptor has been previously reported [14,15,16]. Briefly, the 5-HT6 receptor is a GPCR and is coupled to Gs/adenylyl cyclase pathway (adenylyl cyclase3 isoform) [16,17]. 5-HT6 receptors are uniquely localized in the brain regions, including the olfactory tubercle, cerebral cortex, nucleus accumbens, striatum, hippocampus, thalamus, and cerebellum [12,18], providing an opportunity to identify new treatments modulating functions of specific brain regions. Although peripheral 5-HT6 receptors expression has been reported in rodents, their expression is faint [16]. Thus, selective targeting of 5-HT6 receptors may avoid unwanted side effects arising from other body systems (peripheral as well as other brain regions). In this report, we review the efforts and progress in the drug discovery research for agents targeting the 5-HT6 receptors.

2. Methods

2.1. Literature Search

PubMed (United States National Library of Medicine, Bethesda, MD, USA) and clinical trial registries (ClinicalTrials.gov registry) were searched with terms including, but not limited to, “(serotonin-6 receptor) AND (Clinical trials)”, “(5-HT6 receptor) AND (Clinical trials)”, (5-HT6 receptor antagonist) AND (Clinical trials)”, with publication date restricted to between 1 January 1996 to 30 November 2022. The choice of this search date was based on the cloning of human 5-HT6 receptors in the year 1996. Additionally, searches were performed using internet-based search engines, such as Google and Google Scholar, to manually search for other relevant articles.
The 5-HT6 receptor antagonists that were evaluated in patient populations (clinical trials) were considered and listed by code and compound name (if any). The literature on the non-clinical biology and clinical investigation of these 5-HT6 receptor antagonists were collected from PubMed or ClinicalTrials.gov registry or Google or Google scholar by searching with the term “(compound name) and (code)”, “(compound name) OR (code)”.

2.2. Data Extraction

Information extracted from the clinical literature included indication, NCT number, endpoint and outcome. The information extracted from the non-clinical literature included the code(s), chemical structure, in-vitro profile, and in-vivo profile.

3. Results

The searches resulted in 306 publications. Of these, 87 publications (reports, press releases or documents) were included in this review (Figure 1). Publication references are listed in Supplementary Material.

3.1. 5-HT6 Receptor and Drug Discovery

Initial interest in investigating the potential utility of 5-HT6 receptors for brain disorders arose from the studies suggesting that several clinically used psychotropics (e.g., olanzapine, clozapine, amitriptyline, nortriptyline, to name a few) functioned as 5-HT6 receptors antagonists [12,19]. Drug discovery efforts led to the identification of several selective 5-HT6 receptor agents that act as agonists or antagonists [20,21]. 5-HT6 receptor mediates its effects through other signaling pathways, including the mammalian target of rapamycin pathway [22,23,24,25], cyclin-dependent kinase [25,26,27], Fyn-tyrosine kinase [28], light chain 1 subunit of the microtubule-associated protein 1B [29], and Jun activation domain-binding protein-1 [30]. Research suggests that targeting specific downstream pathways (through functional selectivity) may have unique advantages and can assist in developing treatments with specific therapeutic value [31,32]. Although several 5-HT6 receptor agonists and antagonists with diverse physicochemical properties [20,33,34,35] and specific interaction modes with the receptors have been identified [20,26,27,33,34,35,36], such concepts are yet to be a basis for studies in patient populations.
In animal models, both 5-HT6 receptor agonists and antagonists produced similar behavioral effects [37,38]. Most agents that were progressed to clinical evaluation were 5-HT6 receptor antagonists, and we focus on the therapeutic indications of this class of agents. The chemical structures of these agents are included in Figure 2 and their non-clinical and clinical profiles are included in Table 1, Table 2 and Table 3, respectively.
The initial interest of targeting schizophrenia through 5-HT6 receptors evolved from research indicating a potent affinity of several antipsychotics for the 5-HT6 receptors [8]. In addition, 5-HT6 receptor mRNA was found in the limbic system and striatum, brain regions implicated in the pathogenesis of schizophrenia [12]. A few 5-HT6 receptors antagonists have been studied in schizophrenia patients.
Subsequently, the focus from schizophrenia shifted to AD. Progressive cognitive impairment is the major disease symptom of AD and neuropsychiatric abnormalities are not uncommon [91]. Cholinergic hypofunction is a contributor to memory dysfunction [92]. Considering the involvement of 5-HT6 receptors in the modulation of cholinergic neurotransmission [93], 5-HT6 receptor antagonists were explored as potential treatments for cognitive impairment. Several 5-HT6 receptor antagonists were shown to enhance learning and memory in animal models (for reviews, see [16,42,94,95,96]). Most 5-HT6 receptor antagonists have been studied for potential cognitive benefits in AD patients based on the inferred cognitive benefits from animal models.
Parkinson’s disease dementia (PDD) and dementia with Lewy bodies (DLB) are some of the other neurodegenerative disorders with dementia as a characteristic feature. Deficits/imbalance in the cholinergic neurotransmission has been thought to play a role in the dementias of PDD and DLB [97,98,99]. Considering the localization of 5-HT6 receptors in substantia nigra and cerebral cortex [12,18], and the modulatory role of 5-HT6 receptors on cholinergic neurotransmission, 5-HT6 receptor antagonists may have utility in the treatment of PDD or DLB. Few 5-HT6 receptor antagonists have been studied in PDD or DLB.
Neuropsychiatric symptoms (NPS) are heterogeneous non-cognitive behavioral or mood manifestations of neurodegenerative disorders like AD, PDD and DLB [100,101]. These symptoms include psychosis, agitation/aggression, depression, anxiety, euphoria, apathy, disinhibition, irritability, motor disturbance, night-time behavior, and appetite and eating changes [102,103]. NPS are associated with increased levels of caregiver burden and distress [104,105]. There is no approved treatment for the management of NPS, except pimavanserin for PD psychosis and risperidone for short-term use for behavioral and psychological symptoms of AD (in Europe, Canada, Australia and New Zealand). Considering the affinity of antipsychotics and antidepressants for 5-HT6 receptors and the wide distribution of 5-HT6 receptors in the brain regions implicated for the control of mood and behavior, 5-HT6 agents are candidates for use in the treatment of NPS of AD.
The following section describes the profile of the 5-HT6 receptor antagonists in patient populations.

3.2. Avisetron

In addition to affinity for 5-HT6 receptors, avisetron has affinity for 5-HT2B receptors [39]. In an animal model, avisetron attenuated the effects of apomorphine on the startle prepulse inhibition, suggesting potential effects on the positive symptoms of schizophrenia. Avisetron also attenuated the memory deficits induced by MK-801 in task involving object recognition, passive avoidance, and route memory in the Morris water maze task [39,68] indicating potential effects on the cognitive symptoms of schizophrenia.
Initially the effect of avisetron (4 mg, every day (QD)) was studied in schizophrenia patients receiving stable antipsychotic medication. After treatment for 28 days, those treated with avisetron showed improvement in the PANSS total scores compared to the baseline, whereas no changes were observed with placebo treatment. In addition, significant decreases in PANSS positive subscores were observed with avisetron treatment. Significant changes were also noted in the continuous attention task. Overall, the results were suggestive of beneficial cognitive and anti-psychotic effects of avisetron [69,70].
Subsequently, avisetron was studied for its effects in schizophrenia patients with incomplete remission after receiving stable doses of antipsychotics. The starting dose of avisetron was 4 mg (QD) and was increased to 8 mg (QD) after 1 week. Avisetron treatment showed a nonsignificant trend towards improvement over placebo in PANSS total scores, PANSS positive subscores, and PANSS general psychopathology scale. Overall, avisetron was safe and well-tolerated [70]; however, no further evaluation of avisetron for schizophrenia is reported.

3.3. Cerlapirdine

This agent is a potent 5-HT6 receptor antagonist with an affinity for 5-HT7 and 5-HT2B receptors. Cerlapirdine attenuated scopolamine- and MK-801-induced deficits in the object recognition and contextual fear conditioning tasks. It also attenuated the combined scopolamine and MK-801-induced deficit in the object recognition task. Cerlapirdine was shown to modulate acetylcholine and glutamate in the hippocampus [40].
Based on the pharmacological profile, cerlapirdine (0.5 mg, 1.5 mg, 3 mg and 5 mg, QD) was evaluated in an exploratory study as a monotherapy in mild to moderate AD patients for treatment duration of 4 weeks. Although the primary objective was to assess the safety and tolerability, the efficacy was explored using the MMSE, ADAS-Cog, and subtests of the Cambridge Neuropsychological Test Automated Battery (CANTAB). Trends towards improvement favoring the cerlapirdine treatment were observed on the ADAS-Cog scores and CANTAB [71].
Subsequently, cerlapirdine (1.5 mg, 3 mg and 5 mg, QD) was evaluated in a 52-week phase 2 study for its effects on cognition in mild to moderate AD patients. The primary outcome measure was change from baseline in ADAS-Cog scores at week 24. The study was terminated for reasons of futility (NCT00895895) [72]. Futility may be attributed to low 5-HT6 receptor occupancy i.e., <30% after multiple 5 mg doses [106]. No safety concerns were noted with cerlapirdine in the clinical studies.

3.4. Idalopirdine

In addition to potently blocking the 5-HT6 receptors, idalopirdine has potent affinity for adrenergic receptors (α1A and α1B) and moderate affinity for 5-HT2A and 5-HT2C receptors [41]. In the conditioned avoidance response task, idalopirdine potentiated the effects of haloperidol and risperidone [54] suggesting its potential as an adjunct treatment for schizophrenia. In a rat model, idalopirdine as a standalone treatment attenuated the subchronic phencyclidine-induced cognitive impairment, suggesting its potential utility in the treatment of cognitive impairment associated with schizophrenia [41].
In clinical studies, idalopirdine was evaluated as monotherapy in a small population of 20 schizophrenic patients treated with a dose-escalating regimen of idalopirdine (60/180 mg or 120/240 mg) or placebo. The treatment duration was 14 days, and frequency was once a day. The Brief Assessment of Cognition in Schizophrenia (BACS) was used to assess the changes in cognition. A dose-dependent improvement was observed on BACS, with the effect reaching statistical significance at the highest tested dose of 240 mg. No changes were observed in the placebo treatment arm [73].
Further, idalopirdine was evaluated as an augmentation therapy to risperidone (NCT00810667). Idalopirdine (60 mg, BID) did not show improvement over placebo in schizophrenia symptoms as assessed by Positive and Negative Syndrome Scale (PANSS) total scores at the end of 12 weeks’ treatment. In addition, there were no changes in the BACS scores or the PANSS cognitive subscale scores [74]. Overall, idalopirdine was safe and well-tolerated in schizophrenic patients. No further evaluation of idalopirdine in schizophrenia population has been reported.
Subsequently, idalopirdine was repositioned for the treatment of AD. In animal models, no procognitive-like effects of idalopirdine were observed as a standalone treatment. However, idalopirdine potentiated the effects of donepezil on neuronal oscillations, extracellular brain acetylcholine levels, and blood oxygen level-dependent functional signaling [55,56,57].
Guided by observations in animal models, idalopirdine (30 mg, three times a day (TID)) was studied for its effects on cognition in moderate AD patients as an add-on therapy to donepezil in a phase 2 study. The effect on cognition was assessed based on the change from baseline in ADAS-Cog 11 scores at week 24. A significant improvement in ADAS-Cog 11 scores was observed in the idalopirdine treatment arm compared to placebo. Parallel improvement was also observed in the MMSE scale. A trend towards improvement was observed in functional (ADCS-ADL) and global (Alzheimer’s Disease Cooperative Study Clinical Global Impression of Change (ADCS-CGIC)) outcome measures [75]. Post hoc analysis of the phase 2 study suggested that treatment was associated with improvements in the anxiety and hallucinations domains of the 12-item Neuropsychiatric Inventory scale (NPI-12) [107].
Based on the effects on cognition noted in the phase 2 trial, idalopirdine was evaluated in phase 3 studies as an add-on therapy to donepezil or other cholinesterase inhibitors. The phase 3 studies differed from the phase 2 study, in dosage (10 mg or 30 mg or 60 mg for phase 3 vs. 30 mg for phase 2), and frequency of the idalopirdine treatment (QD for phase 3 vs. TID for phase 2), background therapy (all cholinesterase inhibitors for phase 3 vs. only donepezil for phase 2), cognitive impairment severity (MMSE12-22, mild to moderate for phase 3 vs. MMSE12-19, moderate for phase 2), and study geography (worldwide for phase 3 vs. Europe, Australia, Czech Republic and Canada for phase 2). No effects of idalopirdine were observed on ADAS-Cog 11 scores or other secondary endpoints compared to placebo [76]. In phase 3 studies, the effect of idalopirdine on anxiety was followed-up by selectively studying the anxiety domain of the NPI-12 scale as one of the secondary endpoints. No notable effect was observed (NCT02006641). Overall, idalopirdine was found to be safe and well-tolerated in the clinical studies.

3.5. Intepirdine

This agent has affinity for 5-HT6 and 5-HT2A receptors [42,108]. In animal models, intepirdine enhanced cholinergic neurotransmission, induced neural plasticity, and enhanced cognition [58]. Intepirdine potentiated the effects of donepezil on cholinergic neurotransmission [59].
In AD patients with mild to moderate cognitive deficits, intepirdine (5 mg, 15 mg and 35 mg, QD) was evaluated as a monotherapy for 24 weeks [77]. The effects on global function and cognition were assessed based on the Clinician’s Interview-Based Impression of Change with caregiver input (CIBIC+) and change from baseline in Alzheimer’s Disease Assessment Scale-Cognitive subscale (ADAS-Cog) scores, respectively. Intepirdine treatment over 24 weeks showed improvement in CIBIC+, and the effect reached nominal statistical significance over placebo. No treatment-related effects were observed on the change from baseline in ADAS-Cog scores; however, slope of the dose response from linear trend analysis showed an improvement. Subsequent post hoc analysis suggested beneficial effects of intepirdine on the ADAS-Cog scores in a subgroup of the population with a Mini-Mental State Examination (MMSE) score of <18.
Two trials evaluated the effects of contemporary treatment with intepirdine and donepezil for 24 weeks in mild to moderate AD patients [78,79]. Intepirdine was evaluated at doses of 15 mg and 35 mg (QD) in one study, and another study titrated intepirdine from 15 mg (QD) to 35 mg (QD) at week 4. The co-primary endpoints were change from baseline in CIBIC+ and ADAS-Cog scores. No significant effects of either intepirdine or donepezil were observed on either of the endpoints. The effects of intepirdine were consistent between studies.
Intepirdine (15 mg and 35 mg, QD) was also studied as an add-on therapy to donepezil for 48 weeks in mild to moderate AD patients [79]. The primary endpoints were Clinical Dementia Rating-Sum of Boxes (CDR-SB) and change from baseline on the ADAS-Cog scores at week 24. Treatment with intepirdine was associated with significant improvement in ADAS-Cog scores at week 24 over placebo, and the effects persisted for up to 48 weeks. Parallel improvements were observed in the function as assessed by Alzheimer’s Disease Cooperative Study-Activities of Daily Living (ADCS-ADL).
The observation of potential beneficial effects in earlier studies led to a large phase 3 study evaluating intepirdine (35 mg, QD) as an add-on treatment to donepezil for 24 weeks [80]. The change from baseline in ADAS-Cog scores and ADCS-ADL were co-primary endpoints. No notable effect of intepirdine treatment was observed on either of the endpoints. Intepirdine treatment was associated with a favorable safety profile across all the studies.
The effects of intepirdine (35 mg and 70 mg, QD) were assessed in patients with DLB. The primary efficacy measure was assessment of the effects of intepirdine on motor functions as change from baseline in the Unified Parkinson’s Disease Rating Scale–Part III (UPDRS–III) scores at week 24. Secondary outcome measures assessed the effects on cognition as change from baseline in the ADAS-Cog scores. No significant effects of intepirdine were observed in the change from baseline in the UPDRS–III or ADAS-Cog 11 scores compared to placebo. Intepirdine was well-tolerated, with higher incidences of gastrointestinal adverse events [81].

3.6. Landipirdine

Landipirdine is a potent blocker of both 5-HT6 and 5-HT2A receptors. No reports on the non-clinical profile of landipirdine are publicly available. Landipirdine was evaluated for its effect on cognition in PDD patients as an add-on therapy to cholinesterase inhibitor for 16 weeks. The dose of landipirdine was up-titrated: 20 mg (QD) for week 1, 50 mg (QD) for week 2, and 100 mg (QD) from week 3 through week 16. The effect on cognition was assessed as change from baseline in Cognitive Drug Research Computerized Drug Research Cognition Battery Continuity of Attention (CDRCOA) captured in the ON state. No effects were observed on change from baseline in CDRCOA compared to placebo. Worsening of motor symptoms was observed in those receiving landipirdine [82]. In a post hoc analysis, examination of NPS based on the NPI-12 scale suggested potential improvements in apathy, anxiety, and irritability/lability domains with landipirdine treatment over placebo [109]. No further development of landipirdine has been reported.

3.7. Latrepirdine

This 5-HT6 receptor antagonist has affinities for acetylcholinesterases, N-methyl-D-aspartate receptors, and voltage-gated calcium channels. Many of the effects of latrepirdine were believed to be mediated through the blockade of 5-HT6 receptors [43]. Based on the affinity for 5-HT6 receptors, latrepirdine (20 mg) was studied as an add-on to risperidone therapy in schizophrenic patients transitioning from an acute psychotic episode to symptom remission [83]. No notable differences were observed in the PANSS total or subscale scores between the latrepirdine and placebo treated groups. However, latrepirdine showed statistically significant improvement over the placebo in the 16-item Negative Symptom Assessment (NSA-16) and numerical improvement in cognitive dimensions (working memory, attention, psycho-motor coordination and planning) in comparison with the placebo-treated group. No further studies for latrepirdine in schizophrenic patients have been reported.
In animal models, latrepirdine treatment was associated with memory improvement in a social recognition task [43] and an object recognition task [63]. The interest of developing latrepirdine for AD was based on the outcome from an 8-week open label pilot study. Latrepirdine was evaluated at a dose of 20 mg, TID. The study found improvements in cognition, function, and NPS [44]. An ensuing proof-of-concept study reported similar observations on cognition (assessed based on change from baseline in ADAS-Cog), function (assessed based on change from baseline in ADCS-ADL) and neuropsychiatric symptoms (assessed based on change from baseline in NPI-12). Latrepirdine was evaluated at a dose of 20 mg, TID after 26 weeks of treatment [84]. The effect of latrepirdine on the individual NPS was not reported, except for an absence of effects on dysphoria/depression [44,84]. In the subsequent larger phase 3 studies that were conducted to replicate the earlier findings, no effect was observed on cognition or functions as a standalone treatment for 26 weeks (NCT00675623) or as an add-on treatment to donepezil for 52 weeks (NCT00829374). In the above studies, latrepirdine was evaluated at doses of 5 mg and 20 mg, TID [85]. The effects on NPS were not reported in the results from the phase 3 studies. Latrepirdine was safe and well-tolerated in AD patients.

3.8. Masupirdine

This therapeutic candidate is a potent 5-HT6 receptor antagonist and lacks affinity for other serotonergic receptors subtypes at clinically relevant doses. Masupirdine showed procognitive effects in diverse animal models as standalone and add-on treatment to donepezil. Masupirdine showed beneficial effects on cognition in animals receiving treatment with donepezil and memantine [49,65]. In animal models, masupirdine significantly reduced aggression-like behaviors in the resident–intruder task and decreased dominance levels in the dominant–submissive assay. In addition, masupirdine modulated the cortical dopamine and norepinephrine levels assessed using the brain microdialysis in rats [66].
Contingent on the procognitive effects of masupirdine in combination with donepezil and memantine, a phase 2 study evaluated masupirdine (50 mg and 100 mg, QD) as an add-on treatment to donepezil and memantine in moderate AD patients. The primary efficacy endpoint was the change from baseline in ADAS-Cog scores after 26 weeks of treatment. No notable effects of masupirdine were observed on cognition when compared to the placebo treatment. Overall, masupirdine was safe and well-tolerated in AD patients [86]. A post hoc analysis revealed that masupirdine may have cognitive benefits in patients who are not concurrently treated with memantine high dose (28 mg) [87]. Post hoc observations from the phase 2 study suggested that treatment with masupirdine was associated with improvements in domains of agitation/aggression and psychosis [88].

3.9. SAM-760

It is a potent 5-HT6 receptor antagonist. SAM-760 also acts as an antagonist at 5-HT2A receptors; however, it did not show any significant occupancy of cortical 5-HT2A receptors in humans [110]. SAM-760 showed procognitive effects in diverse animal models including cholinergic and glutamatergic deficit models. It enhanced the cholinergic and glutamatergic neurotransmission in the rat hippocampus and prefrontal cortex [50,51].
SAM-760 was advanced for clinical evaluation to assess its effect on cognition in moderate AD patients with existing NPS. Twelve weeks of treatment with SAM-760 (30 mg, QD) produced no significant effects in the change from baseline in ADAS-Cog scores. No beneficial effects were observed in the secondary outcome measures including NPS as assessed by the NPI-12 scale. Overall, SAM-760 was safe and well-tolerated [90].

4. Conclusions and Outlook

5-HT6 receptors have received considerable attention as potential treatments for cognitive deficits or NPS associated with AD and other neurocognitive disorders. Blocking the functions of 5-HT6 receptors has been the biological effect of agents evaluated in clinical trials. Although the initial focus of 5-HT6 receptors research was intended for the treatment of schizophrenia, most of the advanced clinical studies focus on memory deficits associated with AD or other dementias. However, no conclusive evidence has been observed in clinical trials to support the utility of targeting 5-HT6 receptor antagonists for the treatment of schizophrenia or memory deficits in dementias. The majority of these agents had affinity for other (non-5-HT6) serotonergic receptors, which might partially explain their failure(s).
5-HT6 receptor agents have been shown to have anxiolytic and antidepressant-like properties in animal models [37]. 5-HT6 receptor antagonists also facilitate release of neurotransmitters, such as dopamine and norepinephrine, implicated in mood and behavior [42,59,111]. Based on the affinity of many psychotropic drugs for 5-HT6 receptors, the observations of modulatory role of the receptor on mood, and potential beneficial effects on NPS in patients with dementia, 5-HT6 receptors antagonists may have potential as treatments of NPS in dementia. Many of the 5-HT6 receptor antagonists also blocked the 5-HT2A receptors. Considering the beneficial effects of 5-HT2A receptors blockade in psychiatry, it is essential to understand the role of 5-HT6 receptor on NPS. Characterizing the clinically evaluated agents for their interaction modes with the receptor may further help to delineate agents, and selective 5-HT6 receptor antagonist may target specific downstream pathways and avoid unwanted effects. Among the 5-HT6 receptor antagonists, masupirdine is being evaluated for its effects on agitation in patients with Alzheimer’s-type dementia in a potentially pivotal clinical trial (NCT05397639). The outcome of 5-HT6 receptor antagonists in the past clinical trials has been disappointing. The current trial of masupirdine for agitation will inform future approaches to the clinical utility of selective 5-HT6 receptor antagonists.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biom13020309/s1, Publication references.

Author Contributions

All authors were involved in preparation of manuscript. Authors critically reviewed the manuscript, commented on drafts, and approved the final manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was sponsored by Suven Life Sciences Ltd.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study is available in the Supplementary Materials.

Acknowledgments

JC is supported by NIGMS grant P20GM109025; NINDS grant U01NS093334; NIA grant R01AG053798; NIA grant P20AG068053; NIA grant R35AG71476; and the Disease Drug Discovery Foundation (ADDF).

Conflicts of Interest

Cummings has provided consultation to AB Science, Acadia, Alkahest, AlphaCognition, ALZPathFinder, Annovis, AriBio, Artery, Avanir, Biogen, Biosplice, Cassava, Cerevel, Clinilabs, Cortexyme, Diadem, EIP Pharma, Eisai, GatehouseBio, GemVax, Genentech, Green Valley, Grifols, Janssen, Karuna, Lexeo, Lilly, Lundbeck, LSP, Merck, NervGen, Novo Nordisk, Oligomerix, Ono, Otsuka, PharmacotrophiX, PRODEO, Prothena, ReMYND, Renew, Resverlogix, Roche, Signant Health, Suven, Unlearn AI, Vaxxinity, VigilNeuro pharmaceutical, assessment, and investment companies. Ramakrishna Nirogi, Pradeep Jayarajan, Anil Shinde, Abdul Rasheed Mohammed, Venkata Ramalingayya Grandhi, Vijay Benade, Vinod Kumar Goyal, Renny Abraham, Venkat Jasti are fulltime employees of Suven Life Sciences Ltd.

References

  1. Erspamer, V.; Vialli, M. Ricerche sul secreto delle cellule enterocromaffini. Boll. Soc. Med.-Chir. Pavia 1937, 51, 357–363. [Google Scholar]
  2. Twarog, B.M.; Page, I.H. Serotonin content of some mammalian tissues and urine and a method for its determination. Am. J. Physiol. 1953, 175, 157–161. [Google Scholar] [CrossRef]
  3. Berger, M.; Gray, J.A.; Roth, B.L. The expanded biology of serotonin. Annu. Rev. Med. 2009, 60, 355–366. [Google Scholar] [CrossRef]
  4. Bockaert, J.; Roussignol, G.; Becamel, C.; Gavarini, S.; Joubert, L.; Dumuis, A.; Fagni, L.; Marin, P. GPCR-interacting proteins (GIPs): Nature and functions. Biochem. Soc. Trans. 2004, 32, 851–855. [Google Scholar] [CrossRef]
  5. Millan, M.J.; Marin, P.; Bockaert, J.; Mannoury la Cour, C. signaling at G-protein-coupled serotonin receptors: Recent advances and future research directions. Trends. Pharmacol. Sci. 2008, 29, 454–464. [Google Scholar] [CrossRef]
  6. Kim, D.Y.; Camilleri, M. Serotonin: A mediator of the brain-gut connection. Am. J. Gastroenterol. 2000, 95, 2698–2709. [Google Scholar] [CrossRef]
  7. Charnay, Y.; Léger, L. Brain serotonergic circuitries. Dialogues Clin. Neurosci. 2010, 12, 471–487. [Google Scholar] [CrossRef]
  8. Roth, B.L. Multiple serotonin receptors: Clinical and experimental aspects. Ann. Clin. Psychiatry 1994, 6, 67–78. [Google Scholar] [CrossRef]
  9. Roth, B.L.; Xia, Z. Molecular and cellular mechanisms for the polarized sorting of serotonin receptors: Relevance for genesis and treatment of psychosis. Crit. Rev. Neurobiol. 2004, 16, 229–236. [Google Scholar] [CrossRef]
  10. Pithadia, A.B.; Jain, S.M. 5-Hydroxytryptamine Receptor Subtypes and their Modulators with Therapeutic Potentials. J. Clin. Med. Res. 2009, 1, 72–80. [Google Scholar] [CrossRef]
  11. McCorvy, J.D.; Roth, B.L. Structure and function of serotonin G protein-coupled receptors. Pharmacol. Ther. 2015, 150, 129–142. [Google Scholar] [CrossRef] [Green Version]
  12. Monsma, F.J., Jr.; Shen, Y.; Ward, R.P.; Hamblin, M.W.; Sibley, D.R. Cloning and expression of a novel serotonin receptor with high affinity for tricyclic psychotropic drugs. Mol. Pharmacol. 1993, 43, 320–327. [Google Scholar]
  13. Kohen, R.; Metcalf, M.A.; Khan, N.; Druck, T.; Huebner, K.; Lachowicz, J.E.; Meltzer, H.Y.; Sibley, D.R.; Roth, B.L.; Hamblin, M.W. Cloning, characterization, and chromosomal localization of a human 5-HT6 serotonin receptor. J. Neurochem. 1996, 66, 47–56. [Google Scholar] [CrossRef]
  14. Codony, X.; Burgueño, J.; Ramírez, M.J.; Vela, J.M. 5-HT6 receptor signal transduction second messenger systems. Int. Rev. Neurobiol. 2010, 94, 89–110. [Google Scholar]
  15. Riccioni, T. 5-HT6 receptor characterization. Int. Rev. Neurobiol. 2010, 94, 67–88. [Google Scholar]
  16. Woolley, M.L.; Marsden, C.A.; Fone, K.C. 5-ht6 receptors. Curr. Drug Targets CNS Neurol. Disord. 2004, 3, 59–79. [Google Scholar] [CrossRef]
  17. Brodsky, M.; Lesiak, A.J.; Croicu, A.; Cohenca, N.; Sullivan, J.M.; Neumaier, J.F. 5-HT6 receptor blockade regulates primary cilia morphology in striatal neurons. Brain Res. 2017, 1660, 10–19. [Google Scholar] [CrossRef]
  18. Gérard, C.; Martres, M.P.; Lefèvre, K.; Miquel, M.C.; Vergé, D.; Lanfumey, L.; Doucet, E.; Hamon, M.; el Mestikawy, S. Immuno-localization of serotonin 5-HT6 receptor-like material in the rat central nervous system. Brain Res. 1997, 746, 207–219. [Google Scholar] [CrossRef]
  19. Roth, B.L.; Craigo, S.C.; Choudhary, M.S.; Uluer, A.; Monsma, F.J., Jr.; Shen, Y.; Meltzer, H.Y.; Sibley, D.R. Binding of typical and atypical antipsychotic agents to 5-hydroxytryptamine-6 and 5-hydroxytryptamine-7 receptors. J. Pharmacol. Exp. Ther. 1994, 268, 1403–1410. [Google Scholar]
  20. Bali, A.; Singh, S. Serotonergic 5-HT6 Receptor Antagonists: Heterocyclic Chemistry and Potential Therapeutic Significance. Curr. Top. Med. Chem. 2015, 15, 1643–1662. [Google Scholar] [CrossRef]
  21. Karila, D.; Freret, T.; Bouet, V.; Boulouard, M.; Dallemagne, P.; Rochais, C. Therapeutic Potential of 5-HT6 Receptor Agonists. J. Med. Chem. 2015, 58, 7901–7912. [Google Scholar] [CrossRef] [PubMed]
  22. Meffre, J.; Chaumont-Dubel, S.; Mannoury la Cour, C.; Loiseau, F.; Watson, D.J.; Dekeyne, A.; Séveno, M.; Rivet, J.M.; Gaven, F.; Déléris, P.; et al. 5-HT(6) receptor recruitment of mTOR as a mechanism for perturbed cognition in schizophrenia. EMBO Mol. Med. 2012, 4, 1043–1056. [Google Scholar] [CrossRef]
  23. Bockaert, J.; Bécamel, C.; Chaumont-Dubel, S.; Claeysen, S.; Vandermoere, F.; Marin, P. Novel and atypical pathways for serotonin signaling. Fac. Rev. 2021, 10, 52. [Google Scholar] [CrossRef]
  24. Martin, P.Y.; Doly, S.; Hamieh, A.M.; Chapuy, E.; Canale, V.; Drop, M.; Chaumont-Dubel, S.; Bantreil, X.; Lamaty, F.; Bojarski, A.J.; et al. mTOR activation by constitutively active serotonin6 receptors as new paradigm in neuropathic pain and its treatment. Prog. Neurobiol. 2020, 193, 101846. [Google Scholar] [CrossRef] [PubMed]
  25. Chaumont-Dubel, S.; Dupuy, V.; Bockaert, J.; Bécamel, C.; Marin, P. The 5-HT6 receptor interactome: New insight in receptor signaling and its impact on brain physiology and pathologies. Neuropharmacology 2020, 172, 107839. [Google Scholar] [CrossRef] [PubMed]
  26. Vanda, D.; Canale, V.; Chaumont-Dubel, S.; Kurczab, R.; Satała, G.; Koczurkiewicz-Adamczyk, P.; Krawczyk, M.; Pietruś, W.; Blicharz, K.; Pękala, E.; et al. Imidazopyridine-Based 5-HT6 Receptor Neutral Antagonists: Impact of N1-Benzyl and N1-Phenylsulfonyl Fragments on Different Receptor Conformational States. J. Med. Chem. 2021, 64, 1180–1196. [Google Scholar] [CrossRef]
  27. Drop, M.; Jacquot, F.; Canale, V.; Chaumont-Dubel, S.; Walczak, M.; Satała, G.; Nosalska, K.; Mahoro, G.U.; Słoczyńska, K.; Piska, K.; et al. Neuropathic pain-alleviating activity of novel 5-HT6 receptor inverse agonists derived from 2-aryl-1H-pyrrole-3-carboxamide. Bioorg. Chem. 2021, 115, 105218. [Google Scholar] [CrossRef]
  28. Yun, H.M.; Kim, S.; Kim, H.J.; Kostenis, E.; Kim, J.I.; Seong, J.Y.; Baik, J.H.; Rhim, H. The novel cellular mechanism of human 5-HT6 receptor through an interaction with Fyn. J. Biol. Chem. 2007, 282, 5496–5505. [Google Scholar] [CrossRef]
  29. Kim, S.H.; Kim, D.H.; Lee, K.H.; Im, S.K.; Hur, E.M.; Chung, K.C.; Rhim, H. Direct interaction and functional coupling between human 5-HT6 receptor and the light chain 1 subunit of the microtubule-associated protein 1B (MAP1B-LC1). PLoS ONE 2014, 9, e91402. [Google Scholar] [CrossRef]
  30. Yun, H.M.; Baik, J.H.; Kang, I.; Jin, C.; Rhim, H. Physical interaction of Jab1 with human serotonin 6 G-protein-coupled receptor and their possible roles in cell survival. J. Biol. Chem. 2010, 285, 10016–10029. [Google Scholar] [CrossRef]
  31. Chang, S.D.; Bruchas, M.R. Functional selectivity at GPCRs: New opportunities in psychiatric drug discovery. Neuropsychopharmacology 2014, 39, 248–249. [Google Scholar] [CrossRef]
  32. Kenakin, T. Functional selectivity and biased receptor signaling. J. Pharmacol. Exp. Ther. 2011, 336, 296–302. [Google Scholar] [CrossRef] [PubMed]
  33. Canale, V.; Trybała, W.; Chaumont-Dubel, S.; KoczurkiewiczAdamczyk, P.; Satała, G.; Bento, O.; Blicharz-Futera, K.; Bantreil, X.; Pękala, E.; Bojarski, A.J.; et al. 1-(Arylsulfonyl-isoindol-2- yl)piperazines as 5-HT6R Antagonists: Mechanochemical Synthesis, In Vitro Pharmacological Properties and Glioprotective Activity. Biomolecules 2023, 13, 12. [Google Scholar] [CrossRef] [PubMed]
  34. Kucwaj-Brysz, K.; Baltrukevich, H.; Czarnota, K.; Handzlik, J. Chemical update on the potential for serotonin 5-HT6 and 5-HT7 receptor agents in the treatment of Alzheimer’s disease. Bioorg. Med. Chem. Lett. 2021, 49, 128275. [Google Scholar] [CrossRef] [PubMed]
  35. Liu, K.G.; Robichaud, A.J. 5-HT6 medicinal chemistry. Int. Rev. Neurobiol. 2010, 94, 1–34. [Google Scholar]
  36. Grychowska, K.; Satała, G.; Kos, T.; Partyka, A.; Colacino, E.; Chaumont-Dubel, S.; Bantreil, X.; Wesołowska, A.; Pawłowski, M.; Martinez, J.; et al. Novel 1H-Pyrrolo[3,2-c]quinoline Based 5-HT6 Receptor Antagonists with Potential Application for the Treatment of Cognitive Disorders Associated with Alzheimer’s Disease. ACS Chem. Neurosci. 2016, 7, 972–983. [Google Scholar] [CrossRef]
  37. Wesołowska, A. Potential role of the 5-HT6 receptor in depression and anxiety: An overview of preclinical data. Pharmacol. Rep. 2010, 62, 564–577. [Google Scholar] [CrossRef]
  38. Woods, S.; Clarke, N.N.; Layfield, R.; Fone, K.C. 5-HT(6) receptor agonists and antagonists enhance learning and memory in a conditioned emotion response paradigm by modulation of cholinergic and glutamatergic mechanisms. Br. J. Pharmacol. 2012, 167, 436–449. [Google Scholar] [CrossRef]
  39. Ivachtchenko, A.V.; Lavrovsky, Y.; Ivanenkov, Y.A. AVN-211, Novel and Highly Selective 5-HT6 Receptor Small Molecule Antagonist, for the Treatment of Alzheimer’s Disease. Mol. Pharm. 2016, 13, 945–963. [Google Scholar] [CrossRef]
  40. Comery, T.A.; Aschmies, S.; Haydar, S.; Hughes, Z.; Huselton, C.; Kowal, D.; Kramer, A.; McFarlane, G.; Monaghan, M.; Smith, D.; et al. SAM-531, N,N-dimethyl-3-{[3-(1-naphthylsulfonyl)-1H-indazol-5-yl]oxy} propan-1-amine, a novel serotonin-6 receptor antagonist with preclinical pro-cognitive efficacy. Alzheimers Dement. 2010, 6, S548–S549. [Google Scholar] [CrossRef]
  41. Arnt, J.; Bang-Andersen, B.; Grayson, B.; Bymaster, F.P.; Cohen, M.P.; DeLapp, N.W.; Giethlen, B.; Kreilgaard, M.; McKinzie, D.L.; Neill, J.C.; et al. Lu AE58054, a 5-HT6 antagonist, reverses cognitive impairment induced by subchronic phencyclidine in a novel object recognition test in rats. Int. J. Neuropsychopharmacol. 2010, 13, 1021–1033. [Google Scholar] [CrossRef] [PubMed]
  42. Upton, N.; Chuang, T.T.; Hunter, A.J.; Virley, D.J. 5-HT6 receptor antagonists as novel cognitive enhancing agents for Alzheimer’s disease. Neurotherapeutics 2008, 5, 458–469. [Google Scholar] [CrossRef]
  43. Schaffhauser, H.; Mathiasen, J.R.; Dicamillo, A.; Huffman, M.J.; Lu, L.D.; McKenna, B.A.; Qian, J.; Marino, M.J. Dimebolin is a 5-HT6 antagonist with acute cognition enhancing activities. Biochem. Pharmacol. 2009, 78, 1035–1042. [Google Scholar] [CrossRef] [PubMed]
  44. Bachurin, S.; Bukatina, E.; Lermontova, N.; Tkachenko, S.; Afanasiev, A.; Grigoriev, V.; Grigorieva, I.; Ivanov, Y.; Sablin, S.; Zefirov, N. Antihistamine agent Dimebon as a novel neuroprotector and a cognition enhancer. Ann. N. Y. Acad. Sci. 2001, 939, 425–435. [Google Scholar] [CrossRef] [PubMed]
  45. Wu, J.; Li, Q.; Bezprozvanny, I. Evaluation of Dimebon in cellular model of Huntington’s disease. Mol. Neurodegener. 2008, 3, 15. [Google Scholar] [CrossRef]
  46. Grigorev, V.V.; Dranyi, O.A.; Bachurin, S.O. Comparative study of action mechanisms of dimebon and memantine on AMPA- and NMDA-subtypes glutamate receptors in rat cerebral neurons. Bull. Exp. Biol. Med. 2003, 136, 474–477. [Google Scholar] [CrossRef] [PubMed]
  47. Lermontova, N.N.; Redkozubov, A.E.; Shevtsova, E.F.; Serkova, T.P.; Kireeva, E.G.; Bachurin, S.O. Dimebon and tacrine inhibit neurotoxic action of beta-amyloid in culture and block L-type Ca(2+) channels. Bull. Exp. Biol. Med. 2001, 132, 1079–1083. [Google Scholar] [CrossRef]
  48. Bachurin, S.O.; Shevtsova, E.P.; Kireeva, E.G.; Oxenkrug, G.F.; Sablin, S.O. Mitochondria as a target for neurotoxins and neuroprotective agents. Ann. N. Y. Acad. Sci. 2003, 993, 334–344. [Google Scholar] [CrossRef]
  49. Nirogi, R.; Shinde, A.; Kambhampati, R.S.; Mohammed, A.R.; Saraf, S.K.; Badange, R.K.; Bandyala, T.R.; Bhatta, V.; Bojja, K.; Reballi, V.; et al. Discovery and Development of 1-[(2-Bromophenyl)sulfonyl]-5-methoxy-3-[(4-methyl-1-piperazinyl)methyl]-1H-indole Dimesylate Monohydrate (SUVN-502): A Novel. ; Potent.; Selective and Orally Active Serotonin 6 (5-HT6) Receptor Antagonist for Potential Treatment of Alzheimer’s Disease. J. Med. Chem. 2017, 60, 1843–1859. [Google Scholar] [PubMed]
  50. Bell, J.; Baird-Bellaire, S.; Leil, T.; Comery, T.; Plotka, A.; Antinew, J.; Vandal, G.; Chalon, S.; James, K. Single- and multiple-dose safety, tolerability and pharmacokinetics of a Novel 5HT6 receptor full antagonist (SAM-760) for the treatment of the symptoms of Alzheimer’s disease in healthy young adults and elderly subjects. Alzheimers Dement. 2012, 7, S778. [Google Scholar] [CrossRef]
  51. Comery, T.; Zasadny, K.; Morris, E.; Antinew, J.; Bell, J.; Billing, B.; Boyden, T.; Esterlis, I.; Huang, Y.; Kupiec, J.; et al. Receptor occupancy of the 5-HT6 receptor antagonist SAM-760 in non-human primates and healthy human volunteers. Alzheimers Dement. 2012, 7, S794–S795. [Google Scholar] [CrossRef]
  52. Schmidt, E.; Areberg, J.; Evans, P.; Zann, V.; Søgaard, B. Assessment of the Contribution of CYP2D6 to the Elimination of Idalopirdine as well as the Absolute Bioavailability Following Multiple Oral Dosing. In Proceedings of the ASCPT 2016 Annual Meeting, San Diego, CA, USA, 8–12 March 2016. [Google Scholar]
  53. Galimberti, D.; Scarpini, E. Idalopirdine as a treatment for Alzheimer’s disease. Expert Opin. Investig. Drugs 2015, 24, 981–987. [Google Scholar] [CrossRef]
  54. Arnt, J.; Olsen, C.K. 5-HT6 receptor ligands and their antipsychotic potential. Int. Rev. Neurobiol. 2011, 96, 141–161. [Google Scholar] [PubMed]
  55. Mørk, A.; Russell, R.V.; de Jong, I.E.; Smagin, G. Effects of the 5-HT6 receptor antagonist idalopirdine on extracellular levels of monoamines.; glutamate and acetylcholine in the rat medial prefrontal cortex. Eur. J. Pharmacol. 2017, 799, 1–6. [Google Scholar] [CrossRef] [PubMed]
  56. Ferris, C.F.; Kulkarni, P.; Yee, J.R.; Nedelman, M.; de Jong, I.E.M. The Serotonin Receptor 6 Antagonist Idalopirdine and Acetylcholinesterase Inhibitor Donepezil Have Synergistic Effects on Brain Activity-A Functional MRI Study in the Awake Rat. Front. Pharmacol. 2017, 8, 279. [Google Scholar] [CrossRef]
  57. Amat-Foraster, M.; Leiser, S.C.; Herrik, K.F.; Richard, N.; Agerskov, C.; Bundgaard, C.; Bastlund, J.F.; de Jong, I.E.M. The 5-HT6 receptor antagonist idalopirdine potentiates the effects of donepezil on gamma oscillations in the frontal cortex of anesthetized and awake rats without affecting sleep-wake architecture. Neuropharmacology 2017, 113, 45–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Chuang, A.T.T.; Foley, A.; Pugh, P.L.; Sunter, D.; Tong, X.; Regan, C.; Dawson, L.A.; Medhurst, A.D.; Upton, N. 5-HT6 receptor antagonist SB-742457 as a novel cognitive enhancing agent for Alzheimer’s disease. Alzheimers Dement. 2006, 2, S631–S632. [Google Scholar] [CrossRef]
  59. Dawson, L.A. The central role of 5-HT6 receptors in modulating brain neurochemistry. Int. Rev. Neurobiol. 2011, 96, 1–26. [Google Scholar]
  60. Sabbagh, M.N.; Shill, H.A. Latrepirdine, a potential novel treatment for Alzheimer’s disease and Huntington’s chorea. Curr. Opin. Investig. Drugs 2010, 11, 80–91. [Google Scholar]
  61. Chew, M.L.; Mordenti, J.; Yeoh, T.; Ranade, G.; Qiu, R.; Fang, J.; Liang, Y.; Corrigan, B. Minimization of CYP2D6 Polymorphic Differences and Improved Bioavailability via Transdermal Administration: Latrepirdine Example. Pharm. Res. 2016, 33, 1873–1880. [Google Scholar] [CrossRef]
  62. Santos, J.; Lobato, L.; Vale, N. Clinical pharmacokinetic study of latrepirdine via in silico sublingual administration. Silico Pharmacol. 2021, 9, 29. [Google Scholar] [CrossRef] [PubMed]
  63. Giorgetti, M.; Gibbons, J.A.; Bernales, S.; Alfaro, I.E.; Drieu La Rochelle, C.; Cremers, T.; Altar, C.A.; Wronski, R.; Hutter-Paier, B.; Protter, A.A. Cognition-enhancing properties of Dimebon in a rat novel object recognition task are unlikely to be associated with acetylcholinesterase inhibition or N-methyl-D-aspartate receptor antagonism. J. Pharmacol. Exp. Ther. 2010, 333, 748–757. [Google Scholar] [CrossRef] [PubMed]
  64. Nirogi, R.; Mudigonda, K.; Bhyrapuneni, G.; Muddana, N.R.; Goyal, V.K.; Pandey, S.K.; Palacharla, R.C. Safety, Tolerability and Pharmacokinetics of the Serotonin 5-HT6 Receptor Antagonist, SUVN-502, in Healthy Young Adults and Elderly Subjects. Clin. Drug Investig. 2018, 38, 401–415. [Google Scholar] [CrossRef]
  65. Nirogi, R.; Abraham, R.; Benade, V.; Medapati, R.B.; Jayarajan, P.; Bhyrapuneni, G.; Muddana, N.; Mekala, V.R.; Subramanian, R.; Shinde, A.; et al. SUVN-502, a novel, potent, pure, and orally active 5-HT6 receptor antagonist: Pharmacological, behavioral, and neurochemical characterization. Behav. Pharmacol. 2019, 30, 16–35. [Google Scholar] [CrossRef] [PubMed]
  66. Nirogi, R.; Jayarajan, P.; Abraham, R.; Tadiparthi, J.; Benade, V.; Shinde, A.K.; Mohammed, A.R.; Badange, R.K.; Goyal, V.K.; Palacharla, V.R.C.; et al. Effects of Masupirdine (SUVN-502) on Agitation/Aggression and Psychosis in Patients with Probable Alzheimer’s Disease: A Post Hoc Analysis. Alzheimers Dement. 2022, 18, e062695. [Google Scholar] [CrossRef]
  67. Sawant-Basak, A.; Obach, R.S.; Doran, A.; Lockwood, P.; Schildknegt, K.; Gao, H.; Mancuso, J.; Tse, S.; Comery, T.A. Metabolism of a 5HT6 Antagonist, 2-Methyl-1-(Phenylsulfonyl)-4-(Piperazin-1-yl)-1H-Benzo[d]imidazole (SAM-760): Impact of Sulfonamide Metabolism on Diminution of a Ketoconazole-Mediated Clinical Drug-Drug Interaction. Drug Metab. Dispos. 2018, 46, 934–942. [Google Scholar] [CrossRef]
  68. Morozova, M.A.; Lepilkina, T.A.; Rupchev, G.E.; Beniashvily, A.G.; Burminskiy, D.S.; Potanin, S.S.; Bondarenko, E.V.; Kazey, V.I.; Lavrovsky, Y.; Ivachtchenko, A.V. Add-on clinical effects of selective antagonist of 5HT6 receptors AVN-211 (CD-008-0173) in patients with schizophrenia stabilized on antipsychotic treatment: Pilot study. CNS Spectr. 2014, 19, 316–323. [Google Scholar] [CrossRef]
  69. Avineuro Completed Phase II Clinical Study Of AVN-211, A Selective 5-HT6 Receptor Antagonist. Available online: https://www.biospace.com/article/releases/avineuro-completed-phase-ii-clinical-study-of-avn-211-a-selective-5-ht6-receptor-antagonist-/ (accessed on 1 November 2022).
  70. Morozova, M.; Burminskiy, D.; Rupchev, G.; Lepilkina, T.; Potanin, S.; Beniashvili, A.; Lavrovsky, Y.; Vostokova, N.; Ivaschenko, A. 5-HT6 Receptor Antagonist as an Adjunct Treatment Targeting Residual Symptoms in Patients with Schizophrenia: Unexpected Sex-Related Effects (Double-Blind Placebo-Controlled Trial). J. Clin. Psychopharmacol. 2017, 37, 169–175. [Google Scholar] [CrossRef]
  71. Brisard, C.; Safirstein, B.; Booth, K.; Hua, L.; Brault, Y.; Raje, S.; Leventer, S. Safety, tolerability, and preliminary efficacy of SAM-531, a 5HT-6 antagonist, in subjects with mild-to-moderate Alzheimer’s disease: Results from a phase 2a study. Alzheimers Dement. 2010, 6, S311. [Google Scholar] [CrossRef]
  72. Study Comparing 3 Dosage Levels Of SAM-531 In Outpatients With Mild To Moderate Alzheimer Disease. Available online: https://clinicaltrials.gov/ct2/show/NCT00895895 (accessed on 1 November 2022).
  73. H. Lundbeck A/S, DK. Methods of treating Alzheimer’s disease and pharmaceutical compositions thereof. WO2014037532.
  74. Saegis Pharmaceuticals, Inc. Completes Phase IIa Clinical Study Of SGS518. Available online: https://www.biospace.com/article/releases/saegis-pharmaceuticals-inc-completes-phase-iia-clinical-study-of-sgs518-/ (accessed on 1 November 2022).
  75. Wilkinson, D.; Windfeld, K.; Colding-Jørgensen, E. Safety and efficacy of idalopirdine, a 5-HT6 receptor antagonist, in patients with moderate Alzheimer’s disease (LADDER): A randomised, double-blind, placebo-controlled phase 2 trial. Lancet Neurol. 2014, 13, 1092–1099. [Google Scholar] [CrossRef]
  76. Atri, A.; Frölich, L.; Ballard, C.; Tariot, P.N.; Molinuevo, J.L.; Boneva, N.; Windfeld, K.; Raket, L.L.; Cummings, J.L. Effect of Idalopirdine as Adjunct to Cholinesterase Inhibitors on Change in Cognition in Patients with Alzheimer Disease: Three Randomized Clinical Trials. JAMA 2018, 319, 130–142. [Google Scholar] [CrossRef] [PubMed]
  77. Maher-Edwards, G.; Zvartau-Hind, M.; Hunter, A.J.; Gold, M.; Hopton, G.; Jacobs, G.; Davy, M.; Williams, P. Double-blind, controlled phase II study of a 5-HT6 receptor antagonist, SB-742457, in Alzheimer’s disease. Curr. Alzheimer Res. 2010, 7, 374–385. [Google Scholar] [CrossRef] [PubMed]
  78. Maher-Edwards, G.; Dixon, R.; Hunter, J.; Gold, M.; Hopton, G.; Jacobs, G.; Hunter, J.; Williams, P. SB-742457 and donepezil in Alzheimer disease: A randomized.; placebo-controlled study. Int. J. Geriatr. Psychiatry 2011, 26, 536–544. [Google Scholar] [CrossRef]
  79. Maher-Edwards, G.; Watson, C.; Ascher, J.; Barnett, C.; Boswell, D.; Davies, J.; Fernandez, M.; Kurz, A.; Zanetti, O.; Safirstein, B.; et al. Two randomized controlled trials of SB742457 in mild-to-moderate Alzheimer’s disease. Alzheimers Dement. 2015, 1, 23–36. [Google Scholar] [CrossRef]
  80. Lang, F.M.; Mo, Y.; Sabbagh, M.; Solomon, P.; Boada, M.; Jones, R.W.; Frisoni, G.B.; Grimmer, T.; Dubois, B.; Harnett, M.; et al. Intepirdine as adjunctive therapy to donepezil for mild-to-moderate Alzheimer’s disease: A randomized.; placebo-controlled.; phase 3 clinical trial (MINDSET). Alzheimers Dement. 2021, 7, e12136. [Google Scholar] [CrossRef]
  81. Lang, F.M.; Kwon, D.Y.; Aarsland, D.; Boeve, B.; Tousi, B.; Harnett, M.; Mo, Y.; Noel Sabbagh, M. An international.; randomized.; placebo-controlled.; phase 2b clinical trial of intepirdine for dementia with Lewy bodies (HEADWAY-DLB). Alzheimers Dement. 2021, 7, e12171. [Google Scholar] [CrossRef]
  82. Fernandez, H.H. SYN120 (a dual 5-HT6/5-HT2A antagonist) study to evaluate safety, tolerability, and efficacy in Parkinson’s disease dementia (SYNAPSE): Phase 2a study results. Neurology 2019, 92, S4.005. [Google Scholar]
  83. Morozova, M.A.; Beniashvili, A.G.; Lepilkina, T.A.; Rupchev, G.E. Double-blind placebo-controlled randomized efficacy and safety trial of add-on treatment of dimebon plus risperidone in schizophrenic patients during transition from acute psychotic episode to remission. Psychiatr. Danub. 2012, 24, 159–166. [Google Scholar]
  84. Doody, R.S.; Gavrilova, S.I.; Sano, M.; Thomas, R.G.; Aisen, P.S.; Bachurin, S.O.; Seely, L.; Hung, D.; dimebon investigators. Effect of dimebon on cognition, activities of daily living, behaviour, and global function in patients with mild-to-moderate Alzheimer’s disease: A randomised, double-blind, placebo-controlled study. Lancet 2008, 372, 207–215. [Google Scholar] [CrossRef]
  85. Pfizer And Medivation Announce Results From Two Phase 3 Studies In Dimebon (latrepirdine*) Alzheimer’s Disease Clinical Development Program. Available online: https://www.pfizer.com/news/press-release/press-release-detail/pfizer_and_medivation_announce_results_from_two_phase_3_studies_in_dimebon_latrepirdine_alzheimer_s_disease_clinical_development_program (accessed on 1 November 2022).
  86. Nirogi, R.; Ieni, J.; Goyal, V.K.; Ravula, J.; Jetta, S.; Shinde, A.; Jayarajan, P.; Benade, V.; Palacharla, V.R.C.; Dogiparti, D.K.; et al. Effect of masupirdine (SUVN-502) on cognition in patients with moderate Alzheimer’s disease: A randomized, double-blind, phase 2, proof-of-concept study. Alzheimers Dement. 2022, 8, e12307. [Google Scholar] [CrossRef]
  87. Nirogi, R.; Goyal, V.K.; Benade, V.; Subramanian, R.; Ravula, J.; Jetta, S.; Shinde, A.; Pandey, S.K.; Jayarajan, P.; Jasti, V.; et al. Effect of Concurrent Use of Memantine on the Efficacy of Masupirdine (SUVN-502): A Post Hoc Analysis of a Phase 2 Randomized Placebo-Controlled Study. Neurol. Ther. 2022, 11, 1583–1594. [Google Scholar] [CrossRef] [PubMed]
  88. Nirogi, R.; Jayarajan, P.; Benade, V.; Shinde, A.; Goyal, V.K.; Jetta, S.; Ravula, J.; Abraham, R.; Grandhi, V.R.; Subramanian, R.; et al. Potential beneficial effects of masupirdine (SUVN-502) on agitation/aggression and psychosis in patients with moderate Alzheimer’s disease: Exploratory post hoc analyses. Int. J. Geriatr. Psychiatry 2022, 37, 10.1002/gps.5813. [Google Scholar] [CrossRef]
  89. Masupirdine for the Treatment of Agitation in Dementia of the Alzheimer’s Type. Available online: https://clinicaltrials.gov/ct2/show/NCT05397639 (accessed on 1 November 2022).
  90. Fullerton, T.; Binneman, B.; David, W.; Delnomdedieu, M.; Kupiec, J.; Lockwood, P.; Mancuso, J.; Miceli, J.; Bell, J. A Phase 2 clinical trial of PF-05212377 (SAM-760) in subjects with mild to moderate Alzheimer’s disease with existing neuropsychiatric symptoms on a stable daily dose of donepezil. Alzheimers. Res. Ther. 2018, 10, 38. [Google Scholar] [CrossRef] [PubMed]
  91. Atri, A. The Alzheimer’s Disease Clinical Spectrum: Diagnosis and Management. Med. Clin. N. Am. 2019, 103, 263–293. [Google Scholar] [CrossRef]
  92. Bartus, R.T.; Dean, R.L., 3rd; Beer, B.; Lippa, A.S. The cholinergic hypothesis of geriatric memory dysfunction. Science 1982, 217, 408–414. [Google Scholar] [CrossRef]
  93. Bourson, A.; Borroni, E.; Austin, R.H.; Monsma, F.J., Jr.; Sleight, A.J. Determination of the role of the 5-ht6 receptor in the rat brain: A study using antisense oligonucleotides. J. Pharmacol. Exp. Ther. 1995, 274, 173–180. [Google Scholar] [PubMed]
  94. Mitchell, E.S. 5-HT6 receptor ligands as antidementia drugs. Int. Rev. Neurobiol. 2011, 96, 163–187. [Google Scholar]
  95. Rossé, G.; Schaffhauser, H. 5-HT6 receptor antagonists as potential therapeutics for cognitive impairment. Curr. Top. Med. Chem. 2010, 10, 207–221. [Google Scholar] [CrossRef]
  96. Johnson, C.N.; Ahmed, M.; Miller, N.D. 5-HT6 receptor antagonists: Prospects for the treatment of cognitive disorders including dementia. Curr. Opin. Drug. Discov. Devel. 2008, 11, 642–654. [Google Scholar]
  97. Iarkov, A.; Mendoza, C.; Echeverria, V. Cholinergic Receptor Modulation as a Target for Preventing Dementia in Parkinson’s Disease. Front. Neurosci. 2021, 15, 665820. [Google Scholar] [CrossRef]
  98. Mori, E.; Ikeda, M.; Kosaka, K. Donepezil-DLB Study Investigators. Donepezil for dementia with Lewy bodies: A randomized.; placebo-controlled trial. Ann. Neurol. 2012, 72, 41–52. [Google Scholar] [CrossRef]
  99. Müller, M.L.; Bohnen, N.I. Cholinergic dysfunction in Parkinson’s disease. Curr. Neurol. Neurosci. Rep. 2013, 13, 377. [Google Scholar] [CrossRef] [Green Version]
  100. Assal, F.; Cummings, J.L. Neuropsychiatric symptoms in the dementias. Curr. Opin. Neurol. 2002, 15, 445–450. [Google Scholar] [CrossRef]
  101. Porsteinsson, A.P.; Antonsdottir, I.M. Neuropsychiatric Symptoms in Dementia: A Cause or Consequence? Am. J. Psychiatry 2015, 172, 410–411. [Google Scholar] [CrossRef]
  102. Cerejeira, J.; Lagarto, L.; Mukaetova-Ladinska, E.B. Behavioral and psychological symptoms of dementia. Front. Neurol. 2012, 3, 73. [Google Scholar] [CrossRef]
  103. Radue, R.; Walaszek, A.; Asthana, S. Neuropsychiatric symptoms in dementia. Handb. Clin. Neurol. 2019, 167, 437–454. [Google Scholar]
  104. Khoo, S.A.; Chen, T.Y.; Ang, Y.H.; Yap, P. The impact of neuropsychiatric symptoms on caregiver distress and quality of life in persons with dementia in an Asian tertiary hospital memory clinic. Int. Psychogeriatr. 2013, 25, 1991–1999. [Google Scholar] [CrossRef]
  105. Mukherjee, A.; Biswas, A.; Roy, A.; Biswas, S.; Gangopadhyay, G.; Das, S.K. Behavioural and Psychological Symptoms of Dementia: Correlates and Impact on Caregiver Distress. Dement. Geriatr. Cogn. Dis. Extra 2017, 7, 354–365. [Google Scholar] [CrossRef]
  106. Rogers, J.; Polhamus, D.; Lockwood, P.; Brault, Y.; Desmet, A.; Ito, K.; Romero, K.; Qiu, R.; Corrigan, B.; Gillespie, W.; et al. Model-based analysis to support strategic decision-making: A case study from the development of a 5HT6 antagonist for the treatment of Alzheimer’s disease. Alzheimers Dement. 2012, 8, P585. [Google Scholar] [CrossRef]
  107. Atri, A.; Tong, G.; Isojarvi, J.; Odergren, T. A 5-HT6 antagonist as adjunctive therapy to cholinesterase inhibitors in patients with mild to-moderate Alzheimer’s disease: Idalopirdine in phase III. Am. J. Geriatr. Psychiatry 2016, 24, S147–S148. [Google Scholar] [CrossRef]
  108. Parker, C.A.; Rabiner, E.A.; Gunn, R.N.; Searle, G.; Martarello, L.; Comley, R.A.; Davy, M.; Wilson, A.A.; Houle, S.; Mizrahi, R.; et al. Human Kinetic Modeling of the 5HT6 PET Radioligand 11C-GSK215083 and Its Utility for Determining Occupancy at Both 5HT6 and 5HT2A Receptors by SB742457 as a Potential Therapeutic Mechanism of Action in Alzheimer Disease. J. Nucl. Med. 2015, 56, 1901–1909. [Google Scholar] [CrossRef]
  109. Weintraub, D. Impact of SYN120 (a Dual 5-HT6/5-HT2A antagonist) on psychiatric symptoms in patients with Parkinson’s disease dementia: Results from a Phase 2a Study. Mov. Disord. 2018, 33, S105. [Google Scholar]
  110. Lockwood, P.; Bell, J.; Chen, I.; Miceli, J.; Macci, K.; Van Winkle, J.; Planeta, B.; Henry, S.; Nabulsi, N.; Carson, R. 5HT2a receptor occupancy (RO) in healthy subjects determined by positron emission tomography (PET) following single-dose administration of SAM760 (PF-05212377). Clin. Pharmacol. Ther. 2015, 97, S21. [Google Scholar]
  111. Lacroix, L.P.; Dawson, L.A.; Hagan, J.J.; Heidbreder, C.A. 5-HT6 receptor antagonist SB-271046 enhances extracellular levels of monoamines in the rat medial prefrontal cortex. Synapse 2004, 51, 158–164. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram.
Figure 1. PRISMA flow diagram.
Biomolecules 13 00309 g001
Figure 2. Clinical candidates targeting 5-HT6 receptors.
Figure 2. Clinical candidates targeting 5-HT6 receptors.
Biomolecules 13 00309 g002
Table 1. In-vitro Profile of 5-HT6 Receptor Antagonists in Clinical Development.
Table 1. In-vitro Profile of 5-HT6 Receptor Antagonists in Clinical Development.
AgentsIn-Vitro Profile
5-HT6 Receptors Other Serotonergic ReceptorsOther Receptors
Avisetron
(AVN-211)
Ki 1.09 nM
Kb 0.83–1.97 nM [39]
5-HT1A IC50 > 1000 nM;
5-HT1B IC50 > 1000 nM;
5-HT2A IC50 > 1000 nM;
5-HT2B IC50 96 nM;
5-HT2C IC50 > 1000 nM;
5-HT3 IC50 > 1000 nM;
5-HT4 IC50 > 1000 nM;
5-HT7 IC50 >1000 nM
[39]
5000-fold selectivity over 65 other receptors, enzymes, and ion channels [39]
Cerlapirdine
(SAM-531 or WAY-262531)
Ki 1.3 nM [40]5-HT2B Ki 440 nM;
5-HT7 Ki 881 nM;
5-HT1B IC50 > 1000 nM;
5-HT1D IC50 > 1000 nM;
5-HT2A IC50 > 1000 nM;
5-HT2C IC50 > 1000 nM [40]
Not reported
Idalopirdine
(LY 483518 or SGS518 or Lu AE58054)
Ki 0.83 nM;
EC50 25 nM;
Kb 4.9 nM [41]
5-HT1A Ki 2300 nM;
5-HT1B Ki > 10,000 nM;
5-HT1D Ki 2600 nM;
5-HT1E Ki > 4600 nM;
5-HT1F Ki 2400 nM;
5-HT2A Ki 83 nM;
5-HT2B Ki > 4100 nM;
5-HT2C Ki 250 nM;
5-HT3 IC50 > 10,000 nM;
5-HT4e IC50 > 10,000 nM;
5-HT7 Ki > 10,000 nM [41]
>50-fold selectivity over 100 other receptors, enzymes, and ion channels, except for adrenergic receptors (α1A Ki 21 nM; α1B Ki 22 nM) [41]
Intepirdine
(SB-742457 or RVT-101)
Ki 0.23 nM [42]5-HT2A Ki 10 nM [42] >100-fold selectivity over other receptors, enzymes, and ion channels [42]
Landipirdine
(SYN-120)
Not reported
Latrepirdine
(Dimebon)
Ki 26 nM;
Kb 26 nM [43]
Not reportedWeak inhibitor of cholinesterase, N-methyl-D-aspartate and voltage-gated calcium channels and weak modulator of the mitochondrial permeability transition pore [44,45,46,47,48]
Masupirdine
(SUVN-502)
Ki 2.04 nM
Kb 2.6 nM [49]
5-HT1A Ki 7020 nM;
5-HT1B IC50 > 10,000 nM;
5-HT1D IC50 > 10,000 nM;
5-HT2A Ki 2514 nM;
5-HT2C Ki > 1000 nM;
5-HT4B Ki 4166 nM;
5-HT5A IC50 > 10,000 nM;
5-HT7 IC50 > 10,000 nM [49]
>500-fold selectivity over 100 other targets that includes receptors, ion channels, enzymes, peptides, growth factors, steroids, immunological factors, second messengers, and prostaglandins except for dopamine receptor (D3 Ki 616 nM) and adrenergic receptors (α2A Ki 2570 nM; α2C Kb 619 nM) [49]
SAM-760
(WYE-103760 or PF-05212377)
Ki 0.53 nM;
IC50 0.76 nM [50,51]
Not reported
Table 2. Metabolic and In-vivo Profile of 5-HT6 Receptor Antagonists Recently in Development.
Table 2. Metabolic and In-vivo Profile of 5-HT6 Receptor Antagonists Recently in Development.
AgentsOral
Bioavailability (%)
CYP Isoform Involved in the MetabolismActive MetaboliteDrug Interaction LiabilityIn-Vivo Profile
Avisetron5.73 [39]Not reportedInhibitor of CYP 2B6, 2C9, 2C19 [39]Attenuated the memory deficits induced by MK-801, and scopolamine in the object recognition task, passive avoidance task, and Morris water maze task [39]
Cerlapirdine24 [40]Not reportedNone [40]Attenuated the memory deficits induced by MK-801, scopolamine, combined scopolamine and MK-801 treatment in the object recognition task [40]
Idalopirdine60 # [52]CYP3A4 and CYP2D6 [53]Not reportedLow [52]Improved cognition in phencyclidine-challenged rats; modulated dopamine, norepinephrine and glutamate neurotransmitters in brain; potentiated the effects of donepezil on neuronal oscillations, acetylcholine modulation and blood oxygen level-dependent functional signaling [54,55,56,57]
Intepirdine76 [42]Not reportedAttenuated the memory deficits caused by scopolamine in the object recognition task and passive avoidance task and reversed the memory deficit associated with senile dementia; enhanced medial prefrontal cortex cholinergic neurotransmission as standalone and add-on treatment to donepezil [58,59]
LandipirdineNot reported
Latrepirdine53 [60];
5 to 6% in extensive CYP2D6 metabolizers and 45% in poor CYP2D6 metabolizers * [61]
CYP2D6 [62]Not reportedEnhanced memory in the social recognition task and object recognition task [43,63]
Masupirdine24.9 [49]CYP3A4 [64]Yes [64]No $ [64]Attenuated scopolamine, MK-801 and ageing associated memory deficits; potentiated the effects of donepezil on neuronal oscillations, and acetylcholine modulation; potentiated the effects of memantine on acetylcholine modulation; potentiated the effects of donepezil and memantine on cognition in the Morris water maze task, neuronal oscillations, and acetylcholine modulation; reduced aggression-like behaviors in resident intruder task and dominance levels in the dominant–submissive assay; Modulated cortical dopamine and norepinephrine [65,66]
SAM-760Not reportedCYP3A4/5 [67]Not reportedNo [67]Attenuated the memory deficits induced by MK-801, scopolamine, combined scopolamine and MK-801 treatment in the object recognition task [50,51]
# Absolute bioavailability of the idalopirdine immediate release tablet in healthy male subjects; * Estimated oral bioavailability in extensive and poor CYP2D6 metabolizers; $ Based on in-vitro CYP profile.
Table 3. Clinical Profile of 5-HT6 Antagonists Recently in Development.
Table 3. Clinical Profile of 5-HT6 Antagonists Recently in Development.
AgentsIndicationNCT NumberEndpointOutcome
AvisetronSchizophrenia
(Pilot study) [68,69]
Not availableKey Endpoints: PANSS, CATAs augmentation therapy, avisetron treatment-related benefits were observed in PANSS total scores, PANSS positive subscores and CAT
Schizophrenia
(Phase 2 study) [70]
Not availablePrimary: Change from baseline in the PANSS total scores
Other Key Endpoints: Change from baseline in the CGI-S, CGI-I, NSA-16, PSPS, CogFu, BACS and CPT
As augmentation therapy, a trend towards avisetron treatment-related benefits were observed in PANSS total score, PANSS positive subscores and PANSS general psychopathology scale after 6 weeks of treatment; No notable effects on CGI-S, CGI-I, NSA, PSPS, CogFu, BACS and CPT
CerlapirdineAlzheimer’s disease
(Pilot study) [71]
NCT00481520Key Endpoints: MMSE, ADAS-Cog and CANTABTrend towards improvement was observed with cerlapirdine treatment on the ADAS-Cog 11 and CANTAB at the end of 4 weeks
Alzheimer’s disease
(Phase 2 study) [72]
NCT00895895Primary: Change from baseline in the ADAS-Cog 11 total scores
Other Key Endpoints: Change from baseline in the ADCS-CGIC, CANTAB and NPI-12
No beneficial effects of cerlapirdine were observed at the end of 24 weeks on any of the studied endpoints
IdalopirdineSchizophrenia
(Pilot study) [73]
Not availableSafety, tolerability, pharmacokinetics and pharmacodynamics (cognitive changes assessed using BACS)Safe and well-tolerated as standalone treatment for 14 days; Idalopirdine treatment was associated with dose-dependent pattern of improvement in the BACS endpoint; No effect in the placebo treated group
Schizophrenia
(Phase 2 study) [74]
NCT00810667Primary: Change in PANSS total scores
Other Key Endpoints: Neurocognitive performance using the BACS
As augmentation therapy, no change was observed in the PANSS total scores or BACS scores or PANSS cognitive subscale scores as compared to placebo after 12 weeks of treatment
Alzheimer’s disease (Phase 2 study) [75]NCT01019421Primary: Change from baseline in the ADAS-Cog 11
Other Key Endpoints: Change from baseline in the ADCS-ADL, ADCS-CGIC, MMSE and NPI-12
As augmentation therapy, significant improvements in ADAS-Cog 11 scores were observed as compared to placebo after 24 weeks of treatment; Parallel trend towards improvement in ADCS-ADL and ADCS-CGIC; Improvements in anxiety and hallucinations domains of the NPI-12 scale in a post hoc analysis
Alzheimer’s disease (Phase 3 studies) [76]NCT01955161, NCT02006641, and NCT02006654Primary: Change from baseline in the ADAS-Cog
Other Key Endpoints: Change from baseline in the ADCS-ADL, ADCS-CGIC, MMSE and NPI-12
As augmentation therapy, no significant improvements in ADAS-Cog 11 scores as compared to placebo after 24 weeks of treatment; Similar observations were noted in other endpoints
IntepirdineAlzheimer’s disease
(Phase 2 study) [77]
NCT00224497Primary: Change from baseline in the ADAS-Cog 11 scores and CIBIC+
Other Key Endpoints: Change from baseline in the MMSE and NPI-12
Significant improvement in the CIBIC+ and trend in the ADAS-Cog 11 scores was observed with 24 weeks of intepirdine treatment
Alzheimer’s disease
(Phase 2 studies) [78,79]
NCT00348192 and NCT00708552Primary: Change from baseline in the ADAS-Cog 11 scores and CIBIC+
Other Key Endpoints: Change from baseline in the MMSE and ADCS-ADL
No significant effect was observed on the CIBIC+ or ADAS-Cog 11 scores at the end of 24 weeks of intepirdine treatment
Alzheimer’s disease
(Phase 2 study) [79]
NCT00710684Primary: Change from baseline in the ADAS-Cog 11 scores and CDR-SB
Other Key Endpoints: Change from baseline in the MMSE and ADCS-ADL
As an add-on therapy to donepezil, beneficial effects of intepirdine were observed in the ADAS-Cog 11 scores at the end of 24 weeks and the effects were noted up to 48 weeks; No notable effects were observed on the CDR-SB
Alzheimer’s disease
(Phase 3 study) [80]
NCT02585934Primary: Change from baseline in the ADAS-Cog 11 scores and ADCS-ADL
Other Key Endpoints: Change from baseline in the NPI-12
As an add-on therapy to donepezil, no beneficial effects of intepirdine were observed in the ADAS-Cog 11 or ADCS-ADL scores at the end of 24 weeks
Dementia with Lewy bodies
(Phase 2 study) [81]
NCT02669433Primary: Change from baseline in the UPDRS–III total scores
Other Key Endpoints: Change from baseline in the ADAS-Cog 11 and CIBIC+
No beneficial effects of intepirdine were observed in the UPDRS–III total score at the end of 24 weeks
LandipirdineParkinson’s disease dementia
(Phase 2 study) [82]
NCT02258152Primary: Change from baseline in the CDRCOA total scores
Other Key Endpoints: Change from baseline in the ADCS-CGIC, MoCA and NPI-12
No beneficial effects of landipirdine were observed as an add-on treatment to cholinesterase inhibitor after 16 weeks of treatment;
Post hoc analysis suggested beneficial effects on apathy, anxiety, and irritability/lability
LatrepirdineSchizophrenia
(Phase 2 study)
[83]
Not availableKey Endpoints: PANSS, CGI-S and NSA-16As an add-on therapy, no beneficial effect of latrepirdine was observed in the PANSS total or sub scale scores; Latrepirdine showed statistically significant improvement in the NSA-16 scale
Alzheimer’s disease
(Pilot study) [44]
Not availableKey Endpoint: Bukatina scaleTreatment with latrepirdine was associated with improvements in cognitive function and reduction of NPS
Alzheimer’s disease
(Phase 2 study) [84]
NCT00377715Primary: Change from baseline in the ADAS-Cog 11 scores
Other Key Endpoints: Change from baseline in the MMSE, ADCS-ADL and NPI-12
Significant improvement was observed on the ADAS-Cog 11, MMSE, ADCS-ADL and NPI-12 after 24 weeks of treatment
Alzheimer’s disease
(Phase 3 studies) [85]
NCT00675623 and NCT00829374Primary: Change from baseline in the ADAS-Cog 11 scores and CIBIC+ (NCT00675623) or change from baseline in the ADAS-Cog 11 scores and ADCS-ADL (NCT00829374)
Other Key Endpoints: Change from baseline in the MMSE, ADCS-ADL and NPI-12
No significant effect of latrepirdine treatment was observed as standalone or add-on to donepezil after 26 or 52 weeks of treatment
MasupirdineAlzheimer’s disease
(Phase 2 study) [86,87,88]
NCT02580305Primary: Change from baseline in the ADAS-Cog 11 total scores
Other Key Endpoints: Change from baseline in the ADCS-ADL, MMSE, CDR-SB and NPI-12
No beneficial effects of masupirdine were observed as an add-on treatment to donepezil and memantine in the ADAS-Cog 11 after 26 weeks of treatment; Post hoc analysis suggested potential impact of memantine on the efficacy, and potential beneficial effects on agitation/aggression and psychosis
Alzheimer’s disease Agitation
(Potentially pivotal study) [89]
NCT05397639Primary: Change from baseline in the CMAI items scores aligning to the International Psychogeriatric Association agitation criteria domains
Other Key Endpoints: Change from baseline in the modified ADCS-CGI-C, MMSE and ADAS-Cog 11
Study in progress
SAM-760Alzheimer’s disease
(Phase 2 study) [90]
NCT01712074Primary: Change from baseline in the ADAS-Cog 13 total scores
Other Key Endpoints: Change from baseline in the COWAT, CFT and NPI-12
Trial was stopped after a futility analysis; No beneficial effect of SAM-760 treatment was observed after 12 weeks of treatment
ADAS-Cog—Alzheimer’s Disease Assessment Scale-Cognitive subscale; ADAS-Cog 11—Alzheimer’s Disease Assessment Scale-Cognitive subscale 11; ADAS-Cog 13—Alzheimer’s Disease Assessment Scale-Cognitive subscale 13; ADCS-ADL—Alzheimer’s Disease Cooperative Study—Activities of Daily Living; ADCS-CGIC—Alzheimer’s Disease Cooperative Study Clinical Global Impression of Change; BACS—Brief Assessment of Cognition in Schizophrenia; CANTAB—Cambridge Neuropsychological Test Automated Battery; CAT—Continuous Attention Task; CDRCOA—Computerized Drug Research Cognition Battery Continuity of Attention; CDR-SB—Clinical Dementia Rating scale Sum of Boxes; CFT—Category Fluency Test; CGI-I—Clinical Global Impression–Improvement; CGI-S—Clinical Global Impression–Severity; CIBIC+—Clinician’s Interview-Based Impression of Change with caregiver input; CMAI—Cohen-Mansfield Agitation Inventory; CogFu—Scale for Rating Functioning Related to Cognitive Impairment in Schizophrenia; COWAT—Controlled Oral Word Association Test; CPT—Continuous Performance Test; MMSE—Mini-Mental State Examination; MoCA—Montreal Cognitive Assessment. NPI-12—12-item Neuropsychiatric Inventory; NPS—Neuropsychiatric symptoms; NSA-16—16-item Negative Symptom Assessment; PANSS—Positive and Negative Syndrome Scale; PSPS—Personal and Social Performance Scale; UPDRS–III—Unified Parkinson’s Disease Rating Scale–Part III.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nirogi, R.; Jayarajan, P.; Shinde, A.; Mohammed, A.R.; Grandhi, V.R.; Benade, V.; Goyal, V.K.; Abraham, R.; Jasti, V.; Cummings, J. Progress in Investigational Agents Targeting Serotonin-6 Receptors for the Treatment of Brain Disorders. Biomolecules 2023, 13, 309. https://doi.org/10.3390/biom13020309

AMA Style

Nirogi R, Jayarajan P, Shinde A, Mohammed AR, Grandhi VR, Benade V, Goyal VK, Abraham R, Jasti V, Cummings J. Progress in Investigational Agents Targeting Serotonin-6 Receptors for the Treatment of Brain Disorders. Biomolecules. 2023; 13(2):309. https://doi.org/10.3390/biom13020309

Chicago/Turabian Style

Nirogi, Ramakrishna, Pradeep Jayarajan, Anil Shinde, Abdul Rasheed Mohammed, Venkata Ramalingayya Grandhi, Vijay Benade, Vinod Kumar Goyal, Renny Abraham, Venkat Jasti, and Jeffrey Cummings. 2023. "Progress in Investigational Agents Targeting Serotonin-6 Receptors for the Treatment of Brain Disorders" Biomolecules 13, no. 2: 309. https://doi.org/10.3390/biom13020309

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop