Next Article in Journal
Phloretamide Prevent Hepatic and Pancreatic Damage in Diabetic Male Rats by Modulating Nrf2 and NF-κB
Previous Article in Journal
High Fructose Causes More Prominent Liver Steatohepatitis with Leaky Gut Similar to High Glucose Administration in Mice and Attenuation by Lactiplantibacillus plantarum dfa1
Previous Article in Special Issue
The Influence of Serious Games in the Promotion of Healthy Diet and Physical Activity Health: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

5-HT2C Receptor Stimulation in Obesity Treatment: Orthosteric Agonists vs. Allosteric Modulators

by
Edmund Przegaliński
1,*,
Kacper Witek
1,
Karolina Wydra
1,
Jolanta H. Kotlińska
2 and
Małgorzata Filip
1
1
Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343 Krakow, Poland
2
Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki Street 4a, 20-093 Lublin, Poland
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(6), 1449; https://doi.org/10.3390/nu15061449
Submission received: 15 February 2023 / Revised: 9 March 2023 / Accepted: 12 March 2023 / Published: 17 March 2023
(This article belongs to the Special Issue Feature Articles on Obesity and Weight Loss Treatments)

Abstract

:
Obesity is a substantial health and economic issue, and serotonin (5-hydroxytryptamine, 5-HT) is an important neurotransmitter system involved in the regulation of body weight. The 5-HT2C receptors (5-HT2CRs), one of 16 of the 5-HT receptor (5-HTRs) subtypes, play a significant role in food intake and body weight control. In this review, we focused on the 5-HTR agonists, such as fenfluramines, sibutramine, and lorcaserin, which act directly or indirectly at 5-HT2CRs and have been introduced into the clinic as antiobesity medications. Due to their unwanted effects, they were withdrawn from the market. The 5-HT2CR positive allosteric modulators (PAMs) can be potentially safer active drugs than 5-HT2CR agonists. However, more in vivo validation of PAMs is required to fully determine if these drugs will be effective in obesity prevention and antiobesity pharmacology treatment. Methodology strategy: This review focuses on the role of 5-HT2CR agonism in obesity treatment, such as food intake regulation and weight gain. The literature was reviewed according to the review topic. We searched the PubMed and Scopus databases and Multidisciplinary Digital Publishing Institute open-access scientific journals using the following keyword search strategy depending on the chapter phrases: (1) “5-HT2C receptor” AND “food intake”, and (2) “5-HT2C receptor” AND “obesity” AND “respective agonists”, and (3) “5-HT2C receptor” AND “PAM”. We included preclinical studies (only present the weight loss effects) and double-blind, placebo-controlled, randomized clinical trials published since the 1975s (mostly related to antiobesity treatment), and excluded the pay-walled articles. After the search process, the authors selected, carefully screened, and reviewed appropriate papers. In total, 136 articles were included in this review.

Graphical Abstract

1. Introduction

Overweight and obesity are global epidemics affecting more than 2.5 billion people worldwide. Overweight and obesity are related to a body mass index (BMI) >25 kg/m2 and >30 kg/m2, respectively, and result in substantial health and economic problems. Importantly, obesity is associated with several comorbidities, including chronic diseases, such as diabetes (type 2), cardiovascular pathologies (atherosclerosis, hypertension, and myocardial infarction), stroke, and different kinds of cancers. Thus, obesity is responsible for approximately 15% of human mortality [1]. Therefore, respective medications for obesity are important and are still being investigated in several laboratories.
Serotonin (5-hydroxytryptamine, 5-HT) is a key monoaminergic neurotransmitter in the central nervous system. Mammalian 5-HT is synthesized from the amino acid L-tryptophan by tryptophan hydroxylase to 5-hydroxy-L-tryptophan, and finally by aromatic amino acid decarboxylase enzymes. The 5-HT controls numerous physiological functions, such as thermoregulation, motor activity, wakefulness, cognitive function, and mood [2]. The 5-HT plays a crucial role in food intake modulation and body weight control. In fact, manipulations at a presynaptic level to reduce 5-HT function (e.g., after central administration of 5,7-dihydroxytryptamine, which is a 5-HT neurotoxin) or to enhance 5-HT tone (after 5-hydroxytryptophan, which is the precursor of 5-HT or fenfluramine that releases and inhibits the uptake of 5-HT) provoke an increase or decrease in food intake, respectively [3,4,5].
The multiplicity of 5-HT effects is controlled by a family of 5-HT receptor (5-HTRs) classes, 5-HT1-7R [6]. Preclinical studies using 5-HT2CR knockout demonstrated significantly higher food intake and body weight gain [7,8], while several selective or non-selective 5-HT2CR agonists caused anorectic effects [9]. In addition, the 5-HT2CR antagonist can increase weight gain and food intake [10,11]. This suggests that the 5-HT2CR may be considered the principal therapeutic target of antiobesity drugs among 5-HT signaling.
The 5-HT2CRs are seven transmembrane spanning helices G protein-coupled receptors (GPCRs) with three extracellular, three intracellular loops, and an extracellular amino-terminus. The 5-HT2CRs primarily are distributed postsynaptically in the central nervous system and are especially present in the epithelial cells of the choroid plexus, limbic areas, hippocampal regions, the amygdala, and basal ganglia [12]. The latter receptors are located on neuronal cell types expressed acetylcholine, dopamine, and gamma-aminobutyric acid neurotransmitters. The 5-HT2CRs activate the phospholipase Cβ via Gαq/11 protein and are coupled to phospholipase C in neurons and choroid plexus, and their activation leads to a cellular accumulation of inositol 1,4,5-triphosphate and diglyceride. In addition, 5-HT2CRs have also been shown to regulate ion channels and transport processes as well as activate other downstream effectors [6].
Nowadays, when obesity and overweight are epidemics, the effectiveness of medications is necessary and must be investigated. Whereas the 5-HT system plays a crucial role in food intake and body weight control, 5-HT2CRs are one of the therapeutic targets of antiobesity drugs. Our study aimed to show preclinical research and systematize the obesity treatment efficacy of 5-HT2CR agonists in double-blind, placebo-controlled, and randomized clinical trials. Besides, we focused on the future perspective of newly synthesized pharmaceutical molecules, with 5-HT2CR activity modulation, as a potential antiobesity treatment.

2. 5-HT2C Receptor Action in Food Intake Regulation

The hypothalamus is the most important brain region involved in the central control of feeding and energy expenditure. Within the hypothalamus, the arcuate nucleus controls food intake and metabolism [13]. Functionally, the arcuate nucleus neurons are divided into two antagonistic groups that express specific neuropeptides and elicit disparate physiological actions. The first group consists of orexigenic neurons that express neuropeptide Y and agouti-related peptide to stimulate appetite. The second, anorexigenic group of neurons suppresses appetite with pro-opiomelanocortin (POMC) [14,15]. The modulation of feeding behavior via local 5-HT signaling acts mainly by an anorexigenic effect on POMC neurons. They are controlled by excitatory projections from the raphe nuclei 5-HT-reactive neurons stimulate the POMC-expressing arcuate nucleus neurons [16].
Approximately 25% of adult mouse POMC neurons express the 5-HT2CRs, but activation of POMC neurons via their receptor has been shown to account for the anorexigenic effects of several 5-HT drugs [17]. It has been demonstrated that 5-HT2CRs expressed in hypothalamic POMC/cocaine amphetamine-regulated transcript neurons are required to control energy and glucose homeostasis, implicating POMC neurons as the target for the effect of 5-HT2CR agonists on weight-loss induction and improved glycemic control [6,18]. Moreover, 5-HT2CR activation in POMC neurons both induces the Pomc mRNA expression and increases POMC neuronal activity [19]. Notably, the 5-HT2CR knockout phenotype in POMC neurons provokes hyperphagia, sensitizes mice to diet-induced obesity, and directly dysregulates glucose homeostasis [18]. Moreover, selectively reintroducing the 5-HT2CR to POMC neurons in whole-body 5-HT2CR knockout mice is sufficient to completely negate the knockout phenotype [20]. Finally, 5-HT2CRs re-expression solely in POMC mice neurons is sufficient to mediate the effects of 5-HT drugs on food intake [21].
Food intake is also regulated by the reward brain system (the mesocorticolimbic pathway) structures, such as the ventral tegmental area, nucleus accumbens, prefrontal cortex, hippocampus, and amygdala [22,23]. Changes in reward processing are hypothesized to play a crucial role in the onset and maintenance of binge eating. Binge eating disorder is related to recurrent episodes of frequently consuming unusually large amounts of food and feeling unable to stop eating. Binge eating disorder is also related to its frequent comorbidity, obesity, as well as complications of being overweight [24]. Similar to addictive substances, such as cocaine, food activates the ventral tegmental area–nucleus accumbens pathways and stimulates pleasure by increasing the release of dopamine [25,26]. Moreover, a subset of dopamine neurons of the ventral tegmental area has recently been demonstrated as a key stimulator of binge-like eating behavior in mice [27]. At the same time, 5-HT cell bodies, projecting from the dorsal raphe nuclei to the ventral tegmental area and the nucleus accumbens, and a subset of dopamine and gamma-aminobutyric acid neurons in the ventral tegmental area express the 5-HT2CRs [28,29]. Generally, the activation of 5-HT2CRs plays an inhibitory role in the regulation of reward-related behavior by inhibiting release in certain areas of the brain [30,31]. Activation of ventral tegmental area 5-HT2CR-expressing neurons significantly reduces homeostatic feeding in mice [32].

3. 5-HT2C Receptor Agonists in Control over Food Intake Preclinical Research

Functionally, the 5-HTR agonists are drugs that orthosterically bind to one (=selective agonist) or more (=nonselective agonist) subtypes of 5-HTR. Direct or indirect (through additional mechanisms) 5-HTR activation by agonists provides a similar response to the intended activation by endogenous 5-HT (Table 1). In rodents, the 5-HT2CR agonist drugs reduce food intake in a manner consistent with an enhancement of satiety by action in the 5-HT2CR binding site in the brain hypothalamus.

3.1. Piperazine Derivatives

The first reports showing that the stimulation of 5-HT2CRs is responsible for the reduction of food intake were based on experiments performed with piperazine derivatives (m-chlorophenyl piperazine; m-CPP and tri-fluoromethylphenylpiperazine; TFMPP), which are nonselective 5-HT receptor agonists (Table 1). These compounds reduced food intake in rodents, and such an effect did not appear in mice lacking 5-HT2CRs [7] or in rats pretreated with 5-HT2CR antagonists [37]. It has also been shown that piperazine 5-HT receptor agonists (especially m-CPP) reduce hepatic glucose production and insulin tolerance, and both effects are independent of the reduction in food intake [38]. Other authors presented evidence that the effect of m-CPP and other compounds acting via 5-HT2CRs increases satiety, reduces food intake, does not affect energy expenditure, and decreases body weight [39]. It should also be emphasized that hypophagia, an effect depending on the activation of 5-HT2CRs, is also related to an increase in POMC release in the nucleus arcuatus of the hypothalamus and stimulation of melanocortin receptors in the paraventricular nucleus (PVH) [38,40]. In contrast to the above-mentioned piperazine derivatives, several compounds with different chemical structures that have been previously described are selective 5-HT2CR agonists that reduce food intake in rodents. In fact, Org 37684, Ro 60-175, PNU 22394, VER 3323, YM 348, and WAY 163909 have been found to induce hypophagia, which is reduced or blocked by 5-HT2CR antagonists [37,41,42,43,44]. Interestingly, the hypophagia induced by 5-HT2CR agonists seems to be selective. There is a lack of effect on water intake, and tolerance is not induced after chronic administration. Furthermore, some of these compounds (e.g., PNU 22394) appeared to be clinically active [37].

3.2. Fenfluramines

Acting at the presynaptic level, fenfluramine and d-fenfluramine are indirect agonists of 5-HT receptors (Table 1), among which the 5-HT2CR are mostly required for the antiobesity effect. Accordingly, it has been demonstrated that, in 5-HT2CR knockout mice, the anorexic effect of d-fenfluramine was not induced [21,45]. Furthermore, the hypophagic effect of d-fenfluramine was blocked by 5-HT2CR antagonists [39,46]. Preclinical studies show that low (2.5 or 3 mg/kg) and high (10 mg/kg) doses of fenfluramine or d-fenfluramine reduced food intake and decreased body weight in rat models [47,48,49]. Moreover, in the case of cue-induced relapse, d-fenfluramine (3 mg/kg) blocked the reinstatement of lever pressing to food-seeking behavior in male rats [50].

3.3. Sibutramine

Similar to the antiobesity effect of fenfluramines, another selective inhibitor of presynaptic reuptake of the 5-HT and noradrenaline (NA) is β-phenethylamine—sibutramine (Table 1). Sibutramine action has also been suggested to be connected to the activation of 5-HT2CR and enhancement of POMC [51,52]. Sibutramine (3–10 mg/kg) treatment rats showed reduced food intake and decreased body weight [53,54,55,56,57,58]. Interestingly, intraperitoneal injections of sibutramine (0.5–3.0 mg/kg) reduced feeding on a high fat/high sucrose diet across 2-h feeding sessions, but bilateral injections of sibutramine (2.0–10.0 μg) into either the PVN or the medial nucleus accumbens shell increased the food intake of the sweetened fat diet [59].

3.4. Lorcaserin

A series of new 5-HT2CR agonists, as derivatives of 3-benzazepine, were synthesized by Smith et al. [60]. The most potent and selective compound was (1R)-8-chloro-2,3,4,5-tetrahydro-1-methyl-1H-3-benzazepine (APD 356, lorcaserin) (Table 1). The authors performed in vitro (effect on turnover of [3H]phosphoinositol) and in vivo (effect on food intake and body weight after acute or chronic per os administration of lorcaserin) experiments. The results demonstrated its 5-HT2CR selectivity (versus 5-HT2A and 5-HT2B sharing sequence homology) as well as a reduction in food intake after acute administration and reductions in both food intake and body weight after 28 days of treatment. The effect of lorcaserin on food intake has been examined in different animal models of obesity. Some authors used a regular diet [60,61], and other investigators evaluated lorcaserin in rats maintained on a high-fat diet [36,62,63]. Lorcaserin treatment rats (1–2 mg/kg) reduced the percentage of body weight by selective reduction of body fat mass [36,60]. Moreover, Xu et al. performed their experiments using binge-like eating and hunger-driven feeding in mice, where lorcaserin reduced food intake [27]. Lorcaserin also reversed the binge-like feeding observed following stimulation of the nucleus accumbens μ-opioid receptors and blocked nucleus accumbens μ-opioid enhancement of fat intake [64]. The simple antiobesity effect of lorcaserin may be related to the 5-HT2CRs location in brain structures associated with food intake control. It has been shown that the stimulation of such receptors expressed in POMC neurons induces anorexia [20,65], and the stimulation of 5-HT2CRs located on the brain patterning transcription factor single-minded 1 expressing neurons in the PVH increases food intake in mice [66,67].
Other studies showed a reduction in glucose consumption [61] and decreased binge food intake in rats [63] after lorcaserin (1 or 3 mg/kg) treatment. In addition, the above-mentioned antiobesity activity of 5-HT2CR agonists is related to increased satiety and decreased food intake. Such effects of lorcaserin can also be connected with its inhibitory effect on motivation and impulsivity [68]. In some experiments, the effect of lorcaserin on glycemic control has been shown. It reduced the production of glucose and increased the sensitivity to insulin. However, it did not affect the secretion of insulin. The above effects were independent of weight loss [69]. Importantly, single-dose pharmacokinetic studies have shown rapid absorption, high oral bioavailability, and a moderate half-life of lorcaserin [60].

4. Clinical Effects of 5-HT2C Receptor Agonist Drugs

4.1. Fenfluramines

At the beginning of 1970, fenfluramine and d-fenfluramine (an active dextro-rotatory stereoisomer of fenfluramine) were the first antiobesity drugs acting via the 5-HT system to be introduced on the market. In clinical trials, d-fenfluramine showed dose- and time-dependent weight loss (Table 2). In short-term treatment (3–6 months), a higher dose (30–60 mg) of d-fenfluramine led to greater weight loss than low doses (10–15 mg), compared to placebo patients. Long-term treatment (12 months or more) showed greater weight loss in d-fenfluramine patients compared to short-term treatment. Despite the efficacy of fenfluramine or d-fenfluramine in anorexic action, undesirable effects have been observed in obese patients.
In several clinical trials, cardiovascular and heart valve disorders were observed. Echocardiography performed 12 months after the initiation of fenfluramine-phentermine therapy in twenty-four women demonstrated unusual heart valvular morphology and regurgitation in all patients. Moreover, 30% of them also had newly documented pulmonary hypertension [92]. Another study reported that, of 233 obese patients who took appetite suppressants containing d-fenfluramine, 12.7% of them had a prevalence of cardiac valvular insufficiency [93]. Moreover, after the echocardiography of 200 patients with a history of exposure to anorectic medications, significant aortic valve regurgitation was observed in 14% of patients exposed to d-fenfluramine [94].
Other authors showed occurrences of multivalvular disease and pulmonary hypertension after fenfluramine treatment [95,96,97,98]. Nine controlled studies of fenfluramine showed mild or greater aortic valve regurgitation in 9.6% of treated almost 3300 patients compared with 3.9% of 2000 control subjects [99]. Besides the cardiovascular problems, the most common events reported during d-fenfluramine (15 mg twice daily) treatment were diarrhea, asthenia, dry mouth, headache, and tiredness. Side effects were reported significantly more frequently in patients receiving dexfenfluramine 30 mg twice daily than in those receiving 15 mg twice daily [100].
In 1997, fenfluramines were withdrawn from clinical use due to adverse side effects, such as cardiac valvulopathy and pulmonary hypertension. The most likely explanation for fenfluramine-associated valvulopathy is the activation of 5-HT2BRs by norfenfluramine, being a fenfluramine metabolite. Fenfluramines bind weakly to 5-HT2ARs, 5-HT2BRs, and 5-HT2CRs. Norfenfluramine exhibited a high affinity for 5-HT2BRs, and 5-HT2CRs, and a more moderate affinity for 5-HT2ARs [33]. In cells expressing recombinant 5-HT2BRs, norfenfluramine potently stimulated the hydrolysis of inositol phosphates, increased intracellular Ca2+, and activated the mitogen-activated protein kinase cascade, the latter of which has been linked to mitogenic actions of the 5-HT2BRs. This 5-HT2BRs action is important for heart valvulopathy occurrence. Moreover, the level of 5-HT2BRs and 5-HT2ARs transcripts in heart valves was at least 300-fold higher than the levels of the 5-HT2CR transcript, which were barely detectable [101].

4.2. Sibutramine

Sibutramine was approved by the U.S. Food and Drug Administration (FDA) in 1997 for weight loss in the short- and long-term treatment of obesity. Sibutramine has dose- and time- dependent antiobesity effectiveness, as evidenced in clinical studies (Table 2). Compared to placebo-treated patients, higher doses (15–30 mg) of sibutramine result in greater weight loss than low doses (10 mg). Moreover, sibutramine efficacy in reducing body weight in a long-term period (over one year) was greater than in short-term treatment (six months). However, the Sibutramine Cardiovascular Outcomes (SCOUT) trial showed that, at 12 months but not six months of sibutramine treatment, patients with marked lower baseline blood pressure had the tendency to increase [102]. In some other clinical studies, sibutramine did not increase blood pressure in obese patients [103,104]. Besides, patients taking sibutramine experienced a significant increase in heart rate [83,105,106,107,108,109], but without difference in the overall status of the cardiac valves compared to placebo subjects [105,110,111,112]. Otherwise, sibutramine’s most frequently encountered adverse effects were constipation, dry mouth, headache, insomnia, and nausea.
In 2010, the FDA decided to withdraw sibutramine from the market as a consequence of increased primary outcome events (POEs), such as nonfatal myocardial infarction, nonfatal stroke, resuscitation after cardiac arrest, or cardiovascular death. Indeed, the risk of POEs was increased by 16% in the sibutramine treatment group as compared with the placebo [80]. Nevertheless, other results from SCOUTs did not show an overall deleterious effect of treatment but an increase in POEs in subjects with a history of cardiovascular disease and type 2 diabetes [113,114]. Unexpectedly, sibutramine therapy in obese or overweight high-risk patients induced significant mean reductions for all blood lipids [115]. Although sibutramine and its metabolites exhibit low affinity for 5-HT1A-BRs and 5-HT2ARs, these receptors are reported as a possible target for the causation of unwanted cardiovascular effects [116].

4.3. Lorcaserin

In 2012, the FDA approved lorcaserin on the market as a weight loss medication. The results of several clinical studies have shown its antiobesity effect (Table 2). Despite the time of treatment, lorcaserin (10 mg once and twice daily) caused greater weight loss than with a placebo. The most commonly reported adverse effects were headache, nausea, dizziness, and fatigue, but generally, lorcaserin was well-tolerated. In contrast, the unwanted effects of fenfluramines and sibutramine, in randomized, double-blind, placebo-controlled, multinational clinical trials of lorcaserin treatment, were not associated with any increase in cardiovascular events among patients with high cardiovascular risk [88]. Interestingly, lorcaserin treatment for 6–13 months showed an improvement in metabolic and cardiovascular parameters in obese patients [91,117,118]. It has also been reported that this drug decreases the risk of developing diabetes [119]. At the same time, compared to a placebo, lorcaserin did not affect blood pressure, heart rate, or heart valves [120,121,122].
However, the US Food and Drug Administration Adverse Event Reporting System (FAERS) database scrutinization identified that lorcaserin medication was associated with a significantly greater valvular disorder [123]. Only in overweight and obese patients with renal dysfunction, lorcaserin treatment barely increased cardiovascular disease risk [124]. At the same time, the results of several clinical studies have shown its modest antiobesity effects, and lorcaserin’s position on the market slowly declined. Moreover, patients treated with lorcaserin were diagnosed with more frequent occurrences of cancer. Indeed, incidences of malignancies during lorcaserin pharmacotherapy (though not different from the respective placebo groups) were the reason that it was withdrawn as an antiobesity medication in 2020 [125,126,127].

5. Positive-Allosteric Modulators of the 5-HT2CR

The 5-HT2CR is not only an antiobesity drug target, but also a binding site for positive-allosteric modulators (PAMs). PAMs are groups of substances that increase the affinity and/or efficacy of respective receptor ligands (Figure 1). A potential PAM therapeutic role is the ability to potentiate the effect of an endogenous cognate ligand or other probes interacting orthosterically by binding at a distinct, allosteric, receptor recognition site [128]. Thereafter, several new molecules related to 5-HT2CR PAMs were pharmacologically evaluated. Recently, there have been a few papers showing 5-HT2CR PAM to be potentially safer antiobesity drugs. Garcia-Carceles et al. [129] screened a chemical library from Vivia Biotech using the ExviTech platform and validated and synthesized analogues of Compound 5 (VA240). Among 35 synthesized akins, the most interesting was Compound 11 (PAM 11: (N-[(1-benzyl-1H-indol-3-yl)methyl]pyridin-3-amine, VA012). This compound dose-dependently enhanced 5-HT efficacy (highest potentiation—by 35% at 10 µM—of 5-HT-induced inositol monophosphate release in 5-HT2CR HeLa cells), displayed low binding competition with 5-HT or other orthosteric ligands, and did not exhibit significant off-target activities. Importantly, PAM 11 and WAY 161503 (an agonist of 5-HT2CRs used as a reference drug) were administered acutely to reduce food intake in rats, and PAM 11 acted with higher efficacy and with more prolonged action. Moreover, PAM 11-evoked food intake inhibition was not eliminated after pretreatment with the 5-HT2AR antagonist ketanserin. These findings indicate that the effect of PAM 11 was not related to the activation of 5-HT2ARs and suggest that it has no direct action at the orthosteric site of the 5-HT2CR. Furthermore, PAM 11 administered subchronically (seven days) in a restricted food access model also reduced both food intake and body weight gain in rats. Finally, after combined treatment with PAM 11 and sertraline (a 5-HT reuptake inhibitor) administered in a dose-induced mild feeding suppression, potentiation of feeding reduction was demonstrated. This is clear evidence for the allosteric potentiation of 5-HT-induced anorexia, especially 5-HT2CR-induced anorexia [129]. Another study synthesized a series of N-heterocycle imidazole-linked phenyl cyclopropyl methanone molecules. One of three active 5-HT2CR allosteric modulator molecules, Compound 58, showed unique and beneficial dual characteristics. Compound 58 selectively exhibited PAM response at 5-HT2CRs and negative-allosteric modulator at 5-HT2BRs, which is rarely reported for allosteric modulators. From the point of view of 5-HT2CR agonist’s unwanted cardiovascular effects (depending on 5-HT2BRs activation), the negative 5-HT2BRs modulation is a unique property. Besides, at the same dose, both Compound 58 and lorcaserin reduced food intake in rats [130].

6. Conclusions and Perspectives

This paper reviews the preclinical and clinical efficacy of 5-HT2CR agonists in obesity treatment. Moreover, we showed evidence of new therapeutic molecules, based on 5-HT2CR targets, as potential antiobesity drugs.
In preclinical studies, nonselective or more selective 5-HT2CR agonists showed effectiveness in decreasing body weight, reducing food intake, and inhibiting food seeking-behavior. Consequently, 5-HT2CR drugs, such as fenfluramines, sibutramine, and lorcaserin, were approved in the trade as obesity pharmacology medications. Clinical trials confirmed 5-HT2CR agonist’s action in reduced food intake and decreased body weight in patients with BMI >25 kg/m2 or an ideal body weight of 120–180%. However, after a few years, the 5-HT2CR agonist’s position on the market slowly declined, until being withdrawn from the market due to unwanted effects (higher risk of cardiac valvular abnormalities) occurrence. Nonetheless, the above-mentioned drugs have contributed to understanding the mechanisms of 5-HTR action that permitted the design of new selective molecules. Herein, we showed that 5-HT2CR PAM intensifies 5-HT action for reduced food intake and decreased weight loss in a few in vivo studies. Moreover, we presented selective 5-HT2CR PAM without 5-HT2BR-linked cardiac valve affinity that offers a new angle into the pharmacological potential safer in obesity treatment.
Nowadays, more attention is being paid to precision medicine that offers new grounds for obesity prevention and targeting correct treatment based on genetic, pharmacogenomic, and environmental factors [131,132,133]. Recent human studies underline the connectivity between 5-HT availability and BMI as a predictor of obesity treatment success [134]. Furthermore, the loss of 5-HT2CR function due to mutation can predispose humans to obesity. Thus, sequencing genes encoding the 5-HT2CR should be included in diagnostic panels for obesity [135]. Currently, a series of highly specific 5-HT2CR PAM selectively increase in vitro 5-HT efficacy [136]. This elevates the possibility of 5-HT2CR PAM being used in a more directed way as precision medicine.
In conclusion, we underline the role of 5-HT2CR as a crucial therapeutic target in obesity treatment. Further, 5-HT2CR PAM discovery and syntheses are recommended. Furthermore, 5-HT2CR PAM optimization and clinical weight loss validation are necessary for successful antiobesity treatment and to reveal the full therapeutic potential of PAM.

Author Contributions

Conceptualization and supervision, E.P. and M.F.; methodology, K.W. (Kacper Witek); writing—original draft preparation, E.P., K.W. (Kacper Witek), K.W. (Karolina Wydra), J.H.K. and M.F.; writing—review and editing, E.P. and K.W. (Kacper Witek). All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the statutory fund of the Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hurt, R.T.; Mundi, M.S.; Ebbert, J.O. Challenging Obesity, Diabetes, and Addiction: The Potential of Lorcaserin Extended Release. Diabetes Metab. Syndr. Obes. 2018, 11, 469–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Dutton, A.C.; Barnes, N.M. Anti-Obesity Pharmacotherapy: Future Perspectives Utilising 5-HT2C Receptor Agonists. Drug Discov. Today Ther. Strat. 2006, 3, 577–583. [Google Scholar] [CrossRef]
  3. Saller, C.F.; Stricker, E.M. Hyperphagia and Increased Growth in Rats After Intraventricular Injection of 5,7-Dihydroxytryptamine. Science 1976, 191, 385–387. [Google Scholar] [CrossRef] [PubMed]
  4. Blundell, J.E.; Leshem, M.B. The Effect of 5-Hydroxytryptophan on Food Intake and on the Anorexic Action of Amphetamine and Fenfluramine. J Pharm. Pharmacol. 1975, 27, 31–37. [Google Scholar] [CrossRef] [PubMed]
  5. Duhault, J.; Malen, C.; Boulanger, M.; Voisin, C.; Beregi, L.; Schmitt, H. Fenfluramine and 5 Hydroxytryptamine. I: Is Fenfluramine or Norfenfluramine Involved in the Decrease of Brain 5 Hydroxytryptamine? Arzneim.-Forsch./Drug Res. 1975, 25, 1755–1758. [Google Scholar]
  6. Masson, J.; Emerit, M.B.; Hamon, M.; Darmon, M. Serotonergic Signaling: Multiple Effectors and Pleiotropic Effects. Wiley Interdiscip. Rev. Membr. Transp. Signal. 2012, 1, 685–713. [Google Scholar] [CrossRef]
  7. Tecott, L.H.; Sun, L.M.; Akana, S.F.; Strack, A.M.; Lowenstein, D.H.; Dallman, M.F.; Julius, D. Eating Disorder and Epilepsy in Mice Lacking 5-HT2C Serotonin Receptors. Nature 1995, 374, 542–546. [Google Scholar] [CrossRef]
  8. Nonogaki, K.; Nozue, K.; Oka, Y. Hyperphagia Alters Expression of Hypothalamic 5-HT2C and 5-HT1B Receptor Genes and Plasma Des-Acyl Ghrelin Levels in Ay Mice. Endocrinology 2006, 147, 5893–5900. [Google Scholar] [CrossRef] [Green Version]
  9. Bello, N.T.; Liang, N.C. The Use of Serotonergic Drugs to Treat Obesity—Is There Any Hope? Drug Des. Dev. Ther. 2011, 5, 95–109. [Google Scholar] [CrossRef] [Green Version]
  10. Schuhler, S.; Clark, A.; Joseph, W.; Patel, A.; Lehnen, K.; Stratford, E.; Horan, T.L.; Fone, K.C.F.; Ebling, F.J.P.; Schuhler, A. Involvement of 5-HT Receptors in the Regulation of Food Intake in Siberian Hamsters. J. Neuroendocrinol. 2005, 17, 276–285. [Google Scholar] [CrossRef]
  11. Bonhaus, D.W.; Weinhardt, K.K.; Taylor, M.; Desouza, A.; Mcneeley, P.M.; Szczepanski, K.; Fontana, D.J.; Trinh, J.; Rocha, C.L.; Dawson, M.W.; et al. RS-102221: A Novel High Affinity and Selective, 5-HT2C Receptor Antagonist. Neuropharmacology 1997, 36, 621–629. [Google Scholar] [CrossRef] [PubMed]
  12. Wold, E.A.; Wild, C.T.; Cunningham, K.A.; Zhou, J. Targeting the 5-HT2C Receptor in Biological Context and the Current State of 5-HT2C Receptor Ligand Development. Curr. Top. Med. Chem. 2019, 19, 1381–1398. [Google Scholar] [CrossRef] [PubMed]
  13. Myers, M.G.; Olson, D.P. Central Nervous System Control of Metabolism. Nature 2012, 491, 357–363. [Google Scholar] [CrossRef] [PubMed]
  14. Gropp, E.; Shanabrough, M.; Borok, E.; Xu, A.W.; Janoschek, R.; Buch, T.; Plum, L.; Balthasar, N.; Hampel, B.; Waisman, A.; et al. Agouti-Related Peptide–Expressing Neurons Are Mandatory for Feeding. Nat. Neurosci. 2005, 8, 1289–1291. [Google Scholar] [CrossRef]
  15. Balthasar, N.; Dalgaard, L.T.; Lee, C.E.; Yu, J.; Funahashi, H.; Williams, T.; Ferreira, M.; Tang, V.; McGovern, R.A.; Kenny, C.D.; et al. Divergence of Melanocortin Pathways in the Control of Food Intake and Energy Expenditure. Cell 2005, 123, 493–505. [Google Scholar] [CrossRef]
  16. Van Galen, K.A.; Ter Horst, K.W.; Serlie, M.J. Serotonin, Food Intake, and Obesity. Obes. Rev. 2021, 22, e13210. [Google Scholar] [CrossRef]
  17. Doslikova, B.; Garfield, A.S.; Shaw, J.; Evans, M.L.; Burdakov, D.; Billups, B.; Heisler, L.K. 5-HT2C Receptor agonist Anorectic Efficacy Potentiated by 5-HT1B Receptor agonist Coapplication: An Effect Mediated via Increased Proportion of Pro-Opiomelanocortin Neurons Activated. J. Neurosci. 2013, 33, 9800–9804. [Google Scholar] [CrossRef]
  18. Berglund, E.D.; Liu, C.; Sohn, J.W.; Liu, T.; Kim, M.H.; Lee, C.E.; Vianna, C.R.; Williams, K.W.; Xu, Y.; Elmquist, J.K. Serotonin 2C Receptors in Pro-Opiomelanocortin Neurons Regulate Energy and Glucose Homeostasis. J. Clin. Investig. 2013, 123, 5061–5070. [Google Scholar] [CrossRef] [Green Version]
  19. Wyler, S.C.; Lord, C.C.; Lee, S.; Elmquist, J.K.; Liu, C. Serotonergic Control of Metabolic Homeostasis. Front. Cell. Neurosci. 2017, 11, 277. [Google Scholar] [CrossRef] [Green Version]
  20. Roepke, T.A.; Smith, A.W.; Rønnekleiv, O.K.; Kelly, M.J. Serotonin 5-HT2C Receptor-Mediated Inhibition of the M-Current in Hypothalamic POMC Neurons. Am. J. Physiol. Endocrinol. Metab. 2012, 302, E1399–E1406. [Google Scholar] [CrossRef] [Green Version]
  21. Xu, Y.; Jones, J.E.; Kohno, D.; Williams, K.W.; Lee, C.E.; Choi, M.J.; Anderson, J.G.; Heisler, L.K.; Zigman, J.M.; Lowell, B.B.; et al. 5-HT2CRs Expressed by Pro-Opiomelanocortin Neurons Regulate Energy Homeostasis. Neuron 2008, 60, 582–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Alonso-Alonso, M.; Woods, S.C.; Pelchat, M.; Grigson, P.S.; Stice, E.; Farooqi, S.; Khoo, C.S.; Mattes, R.D.; Beauchamp, G.K. Food Reward System: Current Perspectives and Future Research Needs. Nutr. Rev. 2015, 73, 296–307. [Google Scholar] [CrossRef] [Green Version]
  23. Leenaerts, N.; Jongen, D.; Ceccarini, J.; van Oudenhove, L.; Vrieze, E. The Neurobiological Reward System and Binge Eating: A Critical Systematic Review of Neuroimaging Studies. Int. J. Eat. Disord. 2022, 55, 1421–1458. [Google Scholar] [CrossRef] [PubMed]
  24. Amianto, F.; Ottone, L.; Abbate Daga, G.; Fassino, S. Binge-Eating Disorder Diagnosis and Treatment: A Recap in Front of DSM-5. BMC Psychiatry 2015, 15, 70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Cunningham, K.A.; Fox, R.G.; Anastasio, N.C.; Bubar, M.J.; Stutz, S.J.; Moeller, F.G.; Gilbertson, S.R.; Rosenzweig-Lipson, S. Selective Serotonin 5-HT(2C) Receptor Activation Suppresses the Reinforcing Efficacy of Cocaine and Sucrose but Differentially Affects the Incentive-Salience Value of Cocaine- vs. Sucrose-Associated Cues. Neuropharmacology 2011, 61, 513–523. [Google Scholar] [CrossRef] [Green Version]
  26. Price, A.E.; Anastasio, N.C.; Stutz, S.J.; Hommel, J.D.; Cunningham, K.A. Serotonin 5-HT2C Receptor Activation Suppresses Binge Intake and the Reinforcing and Motivational Properties of High-Fat Food. Front. Pharmacol. 2018, 9, 821. [Google Scholar] [CrossRef] [Green Version]
  27. Xu, P.; He, Y.; Cao, X.; Valencia-Torres, L.; Yan, X.; Saito, K.; Wang, C.; Yang, Y.; Hinton, A.; Zhu, L.; et al. Activation of Serotonin 2C Receptors in Dopamine Neurons Inhibits Binge-like Eating in Mice. Biol. Psychiatry 2017, 81, 737–747. [Google Scholar] [CrossRef] [Green Version]
  28. Bubar, M.J.; Cunningham, K.A. Distribution of Serotonin 5-HT2C Receptors in the Ventral Tegmental Area. Neuroscience 2007, 146, 286–297. [Google Scholar] [CrossRef] [Green Version]
  29. Bubar, M.J.; Stutz, S.J.; Cunningham, K.A. 5-HT2C Receptors Localize to Dopamine and GABA Neurons in the Rat Mesoaccumbens Pathway. PLoS ONE 2011, 6, e20508. [Google Scholar] [CrossRef] [Green Version]
  30. De Deurwaerdère, P.; Navailles, S.; Berg, K.A.; Clarke, W.P.; Spampinato, U. Constitutive Activity of the Serotonin2C Receptor Inhibits In Vivo Dopamine Release in the Rat Striatum and Nucleus Accumbens. J. Neurosci. 2004, 24, 3235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Hayes, D.J.; Mosher, T.M.; Greenshaw, A.J. Differential Effects of 5-HT2C Receptor Activation by WAY 161503 on Nicotine-Induced Place Conditioning and Locomotor Activity in Rats. Behav. Brain Res. 2009, 197, 323–330. [Google Scholar] [CrossRef] [PubMed]
  32. Valencia-Torres, L.; Olarte-Sánchez, C.M.; Lyons, D.J.; Georgescu, T.; Greenwald-Yarnell, M.; Myers, M.G.; Bradshaw, C.M.; Heisler, L.K. Activation of Ventral Tegmental Area 5-HT2C Receptors Reduces Incentive Motivation. Neuropsychopharmacology 2017, 42, 1511–1521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Rothman, R.B.; Baumann, M.H.; Savage, J.E.; Rauser, L.; McBride, A.; Hufeisen, S.J.; Roth, B.L. Evidence for Possible Involvement of 5-HT2B Receptors in the Cardiac Valvulopathy Associated With Fenfluramine and Other Serotonergic Medications. Circulation 2000, 102, 2836–2841. [Google Scholar] [CrossRef] [Green Version]
  34. Setola, V.; Dukat, M.; Glennon, R.A.; Roth, B.L. Molecular Determinants for the Interaction of the Valvulopathic Anorexigen Norfenfluramine with the 5-HT2B Receptor. Mol. Pharmacol. 2005, 68, 20–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Frassetto, S.S.; Delia Santa Rubio, Â.; Lopes, J.J.; Pereira, P.; Brum, C.; Khazzaka, M.; Vinagre, A.S. Locomotor and Peripheral Effects of Sibutramine Modulated by 5-HT2 Receptors. Can. J. Physiol. Pharmacol. 2006, 84, 1239–1244. [Google Scholar] [CrossRef]
  36. Thomsen, W.J.; Grottick, A.J.; Menzaghi, F.; Reyes-Saldana, H.; Espitia, S.; Yuskin, D.; Whelan, K.; Martin, M.; Morgan, M.; Chen, W.; et al. Lorcaserin, a Novel Selective Human 5-Hydroxytryptamine2C Agonist: In Vitro and in Vivo Pharmacological Characterization. J. Pharmacol. Exp. Ther. 2008, 325, 577–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Bickerdike, M. 5-HT2C Receptor agonists as Potential Drugs for the Treatment of Obesity. Curr. Top. Med. Chem. 2003, 3, 885–897. [Google Scholar] [CrossRef]
  38. Georgescu, T.; Lyons, D.; Heisler, L.K. Role of Serotonin in Body Weight, Insulin Secretion and Glycaemic Control. J. Neuroendocrinol. 2021, 33, e12960. [Google Scholar] [CrossRef]
  39. Hayashi, A.; Suzuki, M.; Sasamata, M.; Miyata, K. Agonist diversity in 5-HT2C receptor-mediated weight control in rats. Psychopharmacology 2005, 178, 241–249. [Google Scholar] [CrossRef]
  40. Xu, Y.; Jones, J.E.; Lauzon, D.A.; Anderson, J.G.; Balthasar, N.; Heisler, L.K.; Zinn, A.R.; Lowell, B.B.; Elmquist, J.K. A Serotonin and Melanocortin Circuit Mediates D-Fenfluramine Anorexia. J. Neurosci. 2010, 30, 14630–14634. [Google Scholar] [CrossRef] [Green Version]
  41. Martin, J.R.; Bös, M.; Jenck, F.; Moreau, J.L.; Mutel, V.; Sleight, A.J.; Wichmann, J.; Andrews, J.S.; Berendsen, H.H.G.; Broekkamp, C.L.E.; et al. 5-HT2C Receptor Agonists: Pharmacological Characteristics and Therapeutic Potential. J. Pharmacol. Exp. Ther. 1998, 286, 913–924. [Google Scholar] [PubMed]
  42. Kimura, Y.; Hatanaka, K.I.; Naitou, Y.; Maeno, K.; Shimada, I.; Koakutsu, A.; Wanibuchi, F.; Yamaguchi, T. Pharmacological Profile of YM348, a Novel, Potent and Orally Active 5-HT2C Receptor Agonist. Eur. J. Pharmacol. 2004, 483, 37–43. [Google Scholar] [CrossRef] [PubMed]
  43. Vickers, S.P.; Benwell, K.R.; Porter, R.H.; Bickerdike, M.J.; Kennett, G.A.; Dourish, C.T. Comparative Effects of Continuous Infusion of MCPP, Ro 60-0175 and d-Fenfluramine on Food Intake, Water Intake, Body Weight and Locomotor Activity in Rats. Br. J. Pharmacol. 2000, 130, 1305–1314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Dunlop, J.; Sabb, A.L.; Mazandarani, H.; Zhang, J.; Kalgaonker, S.; Shukhina, E.; Sukoff, S.; Vogel, R.L.; Stack, G.; Schechter, L.; et al. WAY-163909 [(7bR, 10aR)-1,2,3,4,8,9,10,10a-Octahydro-7bH-Cyclopenta-[b][1,4]Diazepino[6,7,1hi]Indole], a Novel 5-Hydroxytryptamine 2C Receptor-Selective Agonist with Anorectic Activity. J. Pharmacol. Exp. Ther. 2005, 313, 862–869. [Google Scholar] [CrossRef]
  45. Vickers, S.P.; Clifton, P.G.; Dourish, C.T.; Tecott, L.H. Reduced Satiating Effect of D-Fenfluramine in Serotonin 5-HT(2C) Receptor Mutant Mice. Psychopharmacology 1999, 143, 309–314. [Google Scholar] [CrossRef]
  46. Vickers, S.P.; Dourish, C.T.; Kennett, G.A. Evidence That Hypophagia Induced by D-Fenfluramine and d-Norfenfluramine in the Rat Is Mediated by 5-HT2C Receptors. Neuropharmacology 2001, 41, 200–209. [Google Scholar] [CrossRef]
  47. Fisler, J.S.; Underberger, S.J.; York, D.A.; Bray, G.A. D-Fenfluramine in a Rat Model of Dietary Fat-Induced Obesity. Pharmacol. Biochem. Behav. 1993, 45, 487–493. [Google Scholar] [CrossRef]
  48. Clifton, P.G.; Lee, M.D.; Dourish, C.T. Similarities in the Action of Ro 60-0175, a 5-HT2C Receptor Agonist, and d-Fenfluramine on Feeding Patterns in the Rat. Psychopharmacology 2000, 152, 256–267. [Google Scholar] [CrossRef]
  49. Brindley, D.N.; Hales, P.; Al-Sieni, A.I.I.; Russell, J.C. Sustained Decreases in Weight and Serum Insulin, Glucose, Triacylglycerol and Cholesterol in JCR:LA-Corpulent Rats Treated with D-Fenfluramine. Br. J. Pharmacol. 1992, 105, 679–685. [Google Scholar] [CrossRef] [Green Version]
  50. Pratt, W.E.; Ford, R.T. Systemic Treatment with D-Fenfluramine, but Not Sibutramine, Blocks Cue-Induced Reinstatement of Food-Seeking Behavior in the Rat. Neurosci. Lett. 2013, 556, 232–237. [Google Scholar] [CrossRef] [Green Version]
  51. Burke, L.K.; Doslikova, B.; D’Agostino, G.; Garfield, A.S.; Farooq, G.; Burdakov, D.; Low, M.J.; Rubinstein, M.; Evans, M.L.; Billups, B.; et al. 5-HT Obesity Medication Efficacy via POMC Activation Is Maintained During Aging. Endocrinology 2014, 155, 3732–3738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Higgs, S.; Cooper, A.J.; Barnes, N.M. Reversal of Sibutramine-Induced Anorexia with a Selective 5-HT(2C) Receptor Antagonist. Psychopharmacology 2011, 214, 941–947. [Google Scholar] [CrossRef] [PubMed]
  53. Levin, B.E.; Dunn-Meynell, A.A. Sibutramine Alters the Central Mechanisms Regulating the Defended Body Weight in Diet-Induced Obese Rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2000, 279, R2222–R2228. [Google Scholar] [CrossRef] [Green Version]
  54. Hansen, H.H.; Hansen, G.; Tang-Christensen, M.; Larsen, P.J.; Axel, A.M.D.; Raben, A.; Mikkelsen, J.D. The Novel Triple Monoamine Reuptake Inhibitor Tesofensine Induces Sustained Weight Loss and Improves Glycemic Control in the Diet-Induced Obese Rat: Comparison to Sibutramine and Rimonabant. Eur. J. Pharmacol. 2010, 636, 88–95. [Google Scholar] [CrossRef] [PubMed]
  55. Madsen, A.N.; Hansen, G.; Paulsen, S.J.; Lykkegaard, K.; Tang-Christensen, M.; Hansen, H.S.; Levin, B.E.; Larsen, P.J.; Knudsen, L.B.; Fosgerau, K.; et al. Long-Term Characterization of the Diet-Induced Obese and Diet-Resistant Rat Model: A Polygenetic Rat Model Mimicking the Human Obesity Syndrome. J. Endocrinol. 2010, 206, 287–296. [Google Scholar] [CrossRef] [Green Version]
  56. Hansen, G.; Jelsing, J.; Vrang, N. Effects of Liraglutide and Sibutramine on Food Intake, Palatability, Body Weight and Glucose Tolerance in the Gubra DIO-Rats. Acta Pharmacol. Sin. 2012, 33, 194–200. [Google Scholar] [CrossRef] [Green Version]
  57. Casado, A.; Rodríguez, V.M.; Portillo, M.P.; Macarulla, M.T.; Abecia, L.C.; Echevarría, E.; Casis, L. Sibutramine Decreases Body Weight Gain and Increases Energy Expenditure in Obese Zucker Rats without Changes in NPY and Orexins. Nutr. Neurosci. 2003, 6, 102–111. [Google Scholar] [CrossRef]
  58. Jackson, H.C.; Needham, A.M.; Hutchins, L.J.; Mazurkiewicz, S.E.; Heal, D.J. Comparison of the Effects of Sibutramine and Other Monoamine Reuptake Inhibitors on Food Intake in the Rat. Br. J. Pharmacol. 1997, 121, 1758–1762. [Google Scholar] [CrossRef] [Green Version]
  59. Pratt, W.E.; Connolly, M.E. Contrasting Effects of Systemic and Central Sibutramine Administration on the Intake of a Palatable Diet in the Rat. Neurosci. Lett. 2010, 484, 30–34. [Google Scholar] [CrossRef]
  60. Smith, B.M.; Smith, J.M.; Tsai, J.H.; Schultz, J.A.; Gilson, C.A.; Estrada, S.A.; Chen, R.R.; Park, D.M.; Prieto, E.B.; Gallardo, C.S.; et al. Discovery and Structure-Activity Relationship of (1R)-8-Chloro-2,3,4,5-Tetrahydro-1-Methyl-1H-3-Benzazepine (Lorcaserin), a Selective Serotonin 5-HT2C Receptor Agonist for the Treatment of Obesity. J. Med. Chem. 2008, 51, 305–313. [Google Scholar] [CrossRef]
  61. Higgs, S.; Cooper, A.J.; Barnes, N.M. The 5-HT2C Receptor Agonist, Lorcaserin, and the 5-HT6 Receptor Antagonist, SB-742457, Promote Satiety; A Microstructural Analysis of Feeding Behaviour. Psychopharmacology 2016, 233, 417–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Higgins, G.A.; Desnoyer, J.; van Niekerk, A.; Silenieks, L.B.; Lau, W.; Thevarkunnel, S.; Izhakova, J.; Delannoy, I.A.M.; Fletcher, P.J.; Delay, J.; et al. Characterization of the 5-HT2C Receptor Agonist Lorcaserin on Efficacy and Safety Measures in a Rat Model of Diet-Induced Obesity. Pharmacol. Res. Perspect. 2015, 3, e00084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Price, A.E.; Brehm, V.D.; Hommel, J.D.; Anastasio, N.C.; Cunningham, K.A. Pimavanserin and Lorcaserin Attenuate Measures of Binge Eating in Male Sprague-Dawley Rats. Front. Pharmacol. 2018, 9, 1424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Blumenthal, S.A.; Pratt, W.E. D-Fenfluramine and Lorcaserin Inhibit the Binge-like Feeding Induced by μ-Opioid Receptor Stimulation of the Nucleus Accumbens in the Rat. Neurosci. Lett. 2018, 687, 43–48. [Google Scholar] [CrossRef] [PubMed]
  65. D’Agostino, G.; Lyons, D.; Cristiano, C.; Lettieri, M.; Olarte-Sanchez, C.; Burke, L.K.; Greenwald-Yarnell, M.; Cansell, C.; Doslikova, B.; Georgescu, T.; et al. Nucleus of the Solitary Tract Serotonin 5-HT2C Receptors Modulate Food Intake. Cell Metab. 2018, 28, 619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Zhan, C.; Zhou, J.; Feng, Q.; Zhang, J.E.; Lin, S.; Bao, J.; Wu, P.; Luo, M. Acute and Long-Term Suppression of Feeding Behavior by POMC Neurons in the Brainstem and Hypothalamus, Respectively. J. Neurosci. 2013, 33, 3624. [Google Scholar] [CrossRef] [Green Version]
  67. Kwon, E.; Jo, Y.H. Activation of the ARCPOMC→MeA Projection Reduces Food Intake. Front. Neural Circuits 2020, 14, 595783. [Google Scholar] [CrossRef]
  68. Higgins, G.A.; Zeeb, F.D.; Fletcher, P.J. Role of Impulsivity and Reward in the Anti-Obesity Actions of 5-HT 2C Receptor Agonists. J. Psychopharmacol. 2017, 31, 1403–1418. [Google Scholar] [CrossRef]
  69. Burke, L.K.; Ogunnowo-Bada, E.; Georgescu, T.; Cristiano, C.; de Morentin, P.B.M.; Valencia Torres, L.; D’Agostino, G.; Riches, C.; Heeley, N.; Ruan, Y.; et al. Lorcaserin Improves Glycemic Control via a Melanocortin Neurocircuit. Mol. Metab. 2017, 6, 1092–1102. [Google Scholar] [CrossRef] [Green Version]
  70. Marbury, T.C.; Angelo, J.E.; Michael Gulley, R.; Krosnick, A.; Sugimoto, D.H.; Zellner, S.R. A Placebo-Controlled, Dose-Response Study of Dexfenfluramine in the Treatment of Obese Patients. Curr. Ther. Res. 1996, 57, 663–674. [Google Scholar] [CrossRef]
  71. Lucas, C.P.; Sandage, B.W. Treatment of Obese Patients with Dexfenfluramine: A Multicenter, Placebo-Controlled Study. Am. J. Ther. 1995, 2, 962–967. [Google Scholar] [CrossRef] [PubMed]
  72. Drent, M.L.; Adèr, H.J.; van der Veen, E.A. The Influence of Chronic Administration of the Serotonin Agonist Dexfenfluramine on Responsiveness to Corticotropin Releasing Hormone and Growth Hormone-Releasing Hormone in Moderately Obese People. J. Endocrinol. Investig. 1995, 18, 780–788. [Google Scholar] [CrossRef] [PubMed]
  73. Guy-Grand, B.; Crepaldi, G.; Vre, P.L.; Apfelbaum, M.; Gries, A.; Turner, P. International Trial of Long-Term Dexfenfluramine in Obesity. Lancet 1989, 2, 1142–1145. [Google Scholar] [CrossRef] [PubMed]
  74. Lafreniere, F.; Lambert, L.J.; Rasio, E.; Serri, O. Effects of Dexfenfluramine Treatment on Body Weight and Postprandial Thermogenesis in Obese Subjects. A Double-Blind Placebo-Controlled Study. Int. J. Obes. Relat. Metab. Disord. 1993, 17, 25–30. [Google Scholar] [PubMed]
  75. Mathus-Vliegen, E.M.H.; van de Voorde, K.; Kok, A.M.E.; Res, A.M.A. Dexfenfluramine in the Treatment of Severe Obesity: A Placebo-Controlled Investigation of the Effects on Weight Loss, Cardiovascular Risk Factors, Food Intake and Eating Behaviour. J. Intern. Med. 1992, 232, 119–127. [Google Scholar] [CrossRef]
  76. Weintraub, M.; Rubio, A.; Golik, A.; Byrne, L.; Scheinbaum, M.L. Sibutramine in Weight Control: A Dose-Ranging, Efficacy Study. Clin. Pharmacol. Ther. 1991, 50, 330–337. [Google Scholar] [CrossRef]
  77. Hanotin, C.; Thomas, F.; Jones, S.P.; Leutenegger, E.; Drouin, P. Efficacy and Tolerability of Sibutramine in Obese Patients: A Dose-Ranging Study. Int. J. Obes. 1997, 22, 32–38. [Google Scholar] [CrossRef] [Green Version]
  78. Bray, G.A.; Blackburn, G.L.; Ferguson, J.M.; Greenway, F.L.; Jain, A.K.; Mendel, C.M.; Mendels, J.; Ryan, D.H.; Schwartz, S.L.; Scheinbaum, M.L.; et al. Sibutramine Produces Dose-Related Weight Loss. Obes. Res. 1999, 7, 189–198. [Google Scholar] [CrossRef] [Green Version]
  79. Di Francesco, V.; Sacco, T.; Zamboni, M.; Bissoli, L.; Zoico, E.; Mazzali, G.; Minniti, A.; Salanitri, T.; Cancelli, F.; Bosello, O. Weight Loss and Quality of Life Improvement in Obese Subjects Treated with Sibutramine: A Double-Blind Randomized Multicenter Study. Ann. Nutr. Metab. 2007, 51, 75–81. [Google Scholar] [CrossRef]
  80. James, W.P.T.; Caterson, I.D.; Coutinho, W.; Finer, N.; van Gaal, L.F.; Maggioni, A.P.; Torp-Pedersen, C.; Sharma, A.M.; Shepherd, G.M.; Rode, R.A.; et al. Effect of Sibutramine on Cardiovascular Outcomes in Overweight and Obese Subjects. N. Engl. J. Med. 2010, 363, 905–917. [Google Scholar] [CrossRef] [Green Version]
  81. Berkowitz, R.I.; Fujioka, K.; Daniels, S.R.; Hoppin, A.G.; Owen, S.; Perry, A.C.; Sothern, M.S.; Renz, C.L.; Pirner, M.A.; Walch, J.K.; et al. Effects of Sibutramine Treatment in Obese Adolescents: A Randomized Trial. Ann. Intern. Med. 2006, 145, 81–90. [Google Scholar] [CrossRef] [PubMed]
  82. McNulty, S.J.; Ur, E.; Williams, G. A Randomized Trial of Sibutramine in the Management of Obese Type 2 Diabetic Patients Treated With Metformin. Diabetes Care 2003, 26, 125–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hauner, H.; Meier, M.; Wendland, G.; Kurscheid, T.; Lauterbach, K. Weight Reduction by Sibutramine in Obese Subjects in Primary Care Medicine: The S.A.T. Study. Exp. Clin. Endocrinol. Diabetes 2004, 112, 201–207. [Google Scholar] [CrossRef] [PubMed]
  84. Dujovne, C.A.; Zavoral, J.H.; Rowe, E.; Mendel, C.M. Effects of Sibutramine on Body Weight and Serum Lipids: A Double-Blind, Randomized, Placebo-Controlled Study in 322 Overweight and Obese Patients with Dyslipidemia. Am. Heart J. 2001, 142, 489–497. [Google Scholar] [CrossRef] [Green Version]
  85. Martin, C.K.; Redman, L.M.; Zhang, J.; Sanchez, M.; Anderson, C.M.; Smith, S.R.; Ravussin, E. Lorcaserin, a 5-HT(2C) Receptor Agonist, Reduces Body Weight by Decreasing Energy Intake without Influencing Energy Expenditure. J. Clin. Endocrinol. Metab. 2011, 96, 837–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Smith, S.R.; Prosser, W.A.; Donahue, D.J.; Morgan, M.E.; Anderson, C.M.; Shanahan, W.R. Lorcaserin (APD356), a Selective 5-HT2C Agonist, Reduces Body Weight in Obese Men and Women. Obesity 2009, 17, 494–503. [Google Scholar] [CrossRef] [PubMed]
  87. Shaw Tronieri, J.; Wadden, T.A.; Berkowitz, R.I.; Chao, A.M.; Pearl, R.L.; Alamuddin, N.; Leonard, S.M.; Carvajal, R.; Bakizada, Z.M.; Pinkasavage, E.; et al. A Randomized Trial of Lorcaserin and Lifestyle Counseling for Maintaining Weight Loss Achieved with a Low-Calorie Diet. Obesity 2018, 26, 299–309. [Google Scholar] [CrossRef]
  88. Bohula, E.A.; Wiviott, S.D.; McGuire, D.K.; Inzucchi, S.E.; Kuder, J.; Im, K.; Fanola, C.L.; Qamar, A.; Brown, C.; Budaj, A.; et al. Cardiovascular Safety of Lorcaserin in Overweight or Obese Patients. N. Engl. J. Med. 2018, 379, 1107–1117. [Google Scholar] [CrossRef]
  89. O’Neil, P.M.; Smith, S.R.; Weissman, N.J.; Fidler, M.C.; Sanchez, M.; Zhang, J.; Raether, B.; Anderson, C.M.; Shanahan, W.R. Randomized Placebo-Controlled Clinical Trial of Lorcaserin for Weight Loss in Type 2 Diabetes Mellitus: The BLOOM-DM Study. Obesity 2012, 20, 1426–1436. [Google Scholar] [CrossRef]
  90. Smith, S.R.; Weissman, N.J.; Anderson, C.M.; Sanchez, M.; Chuang, E.; Stubbe, S.; Bays, H.; Shanahan, W.R. Multicenter, Placebo-Controlled Trial of Lorcaserin for Weight Management. N. Engl. J. Med. 2010, 363, 245–256. [Google Scholar] [CrossRef] [Green Version]
  91. Aronne, L.; Shanahan, W.; Fain, R.; Glicklich, A.; Soliman, W.; Li, Y.; Smith, S. Safety and Efficacy of Lorcaserin: A Combined Analysis of the BLOOM and BLOSSOM Trials. Postgrad. Med. 2014, 126, 7–18. [Google Scholar] [CrossRef] [PubMed]
  92. Connolly, H.M.; Crary, J.L.; McGoon, M.D.; Hensrud, D.D.; Edwards, B.S.; Edwards, W.D.; Schaff, H.V. Valvular Heart Disease Associated with Fenfluramine-Phentermine. N. Engl. J. Med. 1997, 337, 141. [Google Scholar] [CrossRef]
  93. Khan, M.A.; Herzog, C.A.; Peter, J.V.S.; Hartley, G.G.; Madlon-Kay, R.; Dick, C.D.; Asinger, R.W.; Vessey, J.T. The Prevalence of Cardiac Valvular Insufficiency Assessed by Transthoracic Echocardiography in Obese Patients Treated with Appetite-Suppressant Drugs. N. Engl. J. Med. 1998, 339, 713–718. [Google Scholar] [CrossRef]
  94. Kancherla, M.K.; Salti, H.I.; Mulderink, T.A.; Parker, M.; Bonow, R.O.; Mehlman, D.J. Echocardiographic Prevalence of Mitral and/or Aortic Regurgitation in Patients Exposed to Either Fenfluramine-Phentermine Combination or to Dexfenfluramine. Am. J. Cardiol. 1999, 84, 1335–1338. [Google Scholar] [CrossRef] [PubMed]
  95. Surapaneni, P.; Vinales, K.L.; Najib, M.Q.; Chaliki, H.P. Valvular Heart Disease with the Use of Fenfluramine-Phentermine. Tex. Heart Inst. J. 2011, 38, 581. [Google Scholar]
  96. McMurray, C.; Bloomfield, P.; Miller, H.C. Irreversible Pulmonary Hypertension after Treatment with Fenfluramine. Br. Med. J. 1986, 292, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Pouwels, H.; Smeets, J.; Cheriex, E.; Wouters, E. Pulmonary Hypertension and Fenfluramine. Eur. Respir. J. 1990, 3, 606–607. [Google Scholar] [CrossRef] [PubMed]
  98. Bang, W.D.; Kim, J.Y.; Yu, H.T.; Cho, S.S.; Jang, J.Y.; Oh, C.M.; Joung, B.; Chang, H.J. Pulmonary Hypertension Associated with Use of Phentermine. Yonsei Med. J. 2010, 51, 971–973. [Google Scholar] [CrossRef] [Green Version]
  99. Schoonjans, A.S.; Marchau, F.; Paelinck, B.P.; Lagae, L.; Gammaitoni, A.; Pringsheim, M.; Keane, M.G.; Ceulemans, B. Cardiovascular Safety of Low-Dose Fenfluramine in Dravet Syndrome: A Review of Its Benefit-Risk Profile in a New Patient Population. Curr. Med. Res. Opin. 2017, 33, 1773–1781. [Google Scholar] [CrossRef] [Green Version]
  100. Davis, R.; Faulds, D. Dexfenfluramine. An Updated Review of Its Therapeutic Use in the Management of Obesity. Drugs 1996, 52, 696–724. [Google Scholar] [CrossRef]
  101. Fitzgerald, L.W.; Burn, T.C.; Brown, B.S.; Patterson, J.P.; Corjay, M.H.; Valentine, P.A.; Sun, J.H.; Link, J.R.; Abbaszade, I.; Hollis, J.M.; et al. Possible Role of Valvular Serotonin 5-HT(2B) Receptors in the Cardiopathy Associated with Fenfluramine. Mol. Pharmacol. 2000, 57, 75–81. [Google Scholar] [PubMed]
  102. Seimon, R.V.; Espinoza, D.; Ivers, L.; Gebski, V.; Finer, N.; Legler, U.F.; Sharma, A.M.; James, W.P.T.; Coutinho, W.; Caterson, I.D. Changes in Body Weight and Blood Pressure: Paradoxical Outcome Events in Overweight and Obese Subjects with Cardiovascular Disease. Int. J. Obes. 2014, 38, 1165–1171. [Google Scholar] [CrossRef] [PubMed]
  103. Daniels, S.R.; Long, B.; Crow, S.; Styne, D.; Sothern, M.; Vargas-Rodriguez, I.; Harris, L.; Walch, J.; Jasinsky, O.; Cwik, K.; et al. Cardiovascular Effects of Sibutramine in the Treatment of Obese Adolescents: Results of a Randomized, Double-Blind, Placebo-Controlled Study. Pediatrics 2007, 120, e147–e157. [Google Scholar] [CrossRef] [PubMed]
  104. Torp-Pedersen, C.; Caterson, I.; Coutinho, W.; Finer, N.; van Gaal, L.; Maggioni, A.; Sharma, A.; Brisco, W.; Deaton, R.; Shepherd, G.; et al. Cardiovascular Responses to Weight Management and Sibutramine in High-Risk Subjects: An Analysis from the SCOUT Trial. Eur. Heart J. 2007, 28, 2915–2923. [Google Scholar] [CrossRef] [PubMed]
  105. Zannad, F.; Gille, B.; Grentzinger, A.; Bruntz, J.F.; Hammadi, M.; Boivin, J.M.; Hanotin, C.; Igau, B.; Drouin, P. Effects of Sibutramine on Ventricular Dimensions and Heart Valves in Obese Patients during Weight Reduction. Am. Heart J. 2002, 144, 508–515. [Google Scholar] [CrossRef]
  106. Gürsoy, A.; Erdoǧan, M.F.; Cin, M.Ö.; Cesur, M.; Başkal, N. Effect of Sibutramine on Blood Pressure in Patients with Obesity and Well-Controlled Hypertension or Normotension. Endocr. Pract. 2005, 11, 308–312. [Google Scholar] [CrossRef]
  107. De Simone, G.; Romano, C.; de Caprio, C.; Contaldo, F.; Salanitri, T.; di Luzio Paparatti, U.; Pasanisi, F. Effects of Sibutramine-Induced Weight Loss on Cardiovascular System in Obese Subjects. Nutr. Metab. Cardiovasc. Dis. 2005, 15, 24–30. [Google Scholar] [CrossRef]
  108. Heusser, K.; Engeli, S.; Tank, J.; Diedrich, A.; Wiesner, S.; Janke, J.; Luft, F.C.; Jordan, J. Sympathetic Vasomotor Tone Determines Blood Pressure Response to Long-Term Sibutramine Treatment. J. Clin. Endocrinol. Metab. 2007, 92, 1560–1563. [Google Scholar] [CrossRef] [Green Version]
  109. Merenich, J.A. The Long-Term Outcomes of Sibutramine Effectiveness on Weight (LOSE Weight) Study: Evaluating the Role of Drug Therapy Within a Weight Management Program in a Group-Model Health Maintenance Organizati. Am. J. Manag. Care 2004, 10, 369–376. [Google Scholar] [CrossRef]
  110. Fanghänel, G.; Cortinas, L.; Sánchez-Reyes, L.; Berber, A. A Clinical Trial of the Use of Sibutramine for the Treatment of Patients Suffering Essential Obesity. Int. J. Obes. Relat. Metab. Disord. 2000, 24, 144–150. [Google Scholar] [CrossRef] [Green Version]
  111. Guven, A.; Koksal, N.; Cetinkaya, A.; Sokmen, G.; Ozdemir, R. Effects of the Sibutramine Therapy on Pulmonary Artery Pressure in Obese Patients. Diabetes Obes. Metab. 2004, 6, 50–55. [Google Scholar] [CrossRef] [PubMed]
  112. Saraç, S.; Saraç, F. Cardiac Valve Evaluation and Adipokine Levels in Obese Women Treated with Sibutramine. Anadolu Kardiyol. Derg. 2010, 10, 226–232. [Google Scholar] [CrossRef] [PubMed]
  113. Caterson, I.D.; Finer, N.; Coutinho, W.; van Gaal, L.F.; Maggioni, A.P.; Torp-Pedersen, C.; Sharma, A.M.; Legler, U.F.; Shepherd, G.M.; Rode, R.A.; et al. Maintained Intentional Weight Loss Reduces Cardiovascular Outcomes: Results from the Sibutramine Cardiovascular OUTcomes (SCOUT) Trial. Diabetes Obes. Metab. 2012, 14, 523–530. [Google Scholar] [CrossRef] [PubMed]
  114. Maggioni, A.P.; Caterson, I.; Coutinho, W.; Finer, N.; van Gaal, L.; Sharma, A.M.; Torp-Pedersen, C.; Bacher, P.; Shepherd, G.; Sun, R.; et al. Tolerability of Sibutramine during a 6-Week Treatment Period in High-Risk Patients with Cardiovascular Disease and/or Diabetes: A Preliminary Analysis of the Sibutramine Cardiovascular Outcomes (SCOUT) Trial. J. Cardiovasc. Pharmacol. 2008, 52, 393–402. [Google Scholar] [CrossRef]
  115. Weeke, P.; Andersson, C.; Fosbøl, E.L.; Brendorp, B.; Køber, L.; Sharma, A.M.; Finer, N.; James, P.T.; Caterson, I.D.; Rode, R.A.; et al. The Weight Lowering Effect of Sibutramine and Its Impact on Serum Lipids in Cardiovascular High Risk Patients with and without Type 2 Diabetes Mellitus—An Analysis from the SCOUT Lead-in Period. BMC Endocr. Disord. 2010, 10, 3. [Google Scholar] [CrossRef] [Green Version]
  116. Nisoli, E.; Carruba, M.O. An Assessment of the Safety and Efficacy of Sibutramine, an Anti-Obesity Drug with a Novel Mechanism of Action. Obes. Rev. 2000, 1, 127–139. [Google Scholar] [CrossRef]
  117. Tuccinardi, D.; Farr, O.M.; Upadhyay, J.; Oussaada, S.M.; Mathew, H.; Paschou, S.A.; Perakakis, N.; Koniaris, A.; Kelesidis, T.; Mantzoros, C.S. Lorcaserin Treatment Decreases Body Weight and Reduces Cardiometabolic Risk Factors in Obese Adults: A Six-Month, Randomized, Placebo-Controlled, Double-Blind Clinical Trial. Diabetes Obes. Metab. 2019, 21, 1487–1492. [Google Scholar] [CrossRef]
  118. Pi-Sunyer, X.; Shanahan, W.; Fain, R.; Ma, T.; Garvey, W.T. Impact of Lorcaserin on Glycemic Control in Overweight and Obese Patients with Type 2 Diabetes: Analysis of Week 52 Responders and Nonresponders. Postgrad. Med. 2016, 128, 591–597. [Google Scholar] [CrossRef]
  119. Bohula, E.A.; Scirica, B.M.; Inzucchi, S.E.; McGuire, D.K.; Keech, A.C.; Smith, S.R.; Kanevsky, E.; Murphy, S.A.; Leiter, L.A.; Dwyer, J.P.; et al. Effect of Lorcaserin on Prevention and Remission of Type 2 Diabetes in Overweight and Obese Patients (CAMELLIA-TIMI 61): A Randomised, Placebo-Controlled Trial. Lancet 2018, 392, 2269–2279. [Google Scholar] [CrossRef]
  120. Fidler, M.C.; Sanchez, M.; Raether, B.; Weissman, N.J.; Smith, S.R.; Shanahan, W.R.; Anderson, C.M. A One-Year Randomized Trial of Lorcaserin for Weight Loss in Obese and Overweight Adults: The BLOSSOM Trial. J. Clin. Endocrinol. Metab. 2011, 96, 3067–3077. [Google Scholar] [CrossRef] [Green Version]
  121. Weissman, N.J.; Sanchez, M.; Koch, G.G.; Smith, S.R.; Shanahan, W.R.; Anderson, C.M. Echocardiographic Assessment of Cardiac Valvular Regurgitation with Lorcaserin from Analysis of 3 Phase 3 Clinical Trials. Circ. Cardiovasc. Imaging 2013, 6, 560–567. [Google Scholar] [CrossRef] [PubMed]
  122. Greenway, F.L.; Shanahan, W.; Fain, R.; Ma, T.; Rubino, D. Safety and Tolerability Review of Lorcaserin in Clinical Trials. Clin. Obes. 2016, 6, 285–295. [Google Scholar] [CrossRef] [PubMed]
  123. Gorelik, E.; Gorelik, B.; Masarwa, R.; Perlman, A.; Hirsh-Raccah, B.; Matok, I. The Cardiovascular Safety of Antiobesity Drugs-Analysis of Signals in the FDA Adverse Event Report System Database. Int. J. Obes. 2020, 44, 1021–1027. [Google Scholar] [CrossRef] [PubMed]
  124. Scirica, B.M.; Bohula, E.A.; Dwyer, J.P.; Qamar, A.; Inzucchi, S.E.; McGuire, D.K.; Keech, A.C.; Smith, S.R.; Murphy, S.A.; Im, K.; et al. Lorcaserin and Renal Outcomes in Obese and Overweight Patients in the CAMELLIA-TIMI 61 Trial. Circulation 2019, 139, 366–375. [Google Scholar] [CrossRef] [PubMed]
  125. Bray, G.A. Medical Treatment of Obesity: The Past, the Present and the Future. Best Pract. Res. Clin. Gastroenterol. 2014, 28, 665–684. [Google Scholar] [CrossRef] [PubMed]
  126. De Andrade Mesquita, L.; Fagundes Piccoli, G.; Richter da Natividade, G.; Frison Spiazzi, B.; Colpani, V.; Gerchman, F. Is Lorcaserin Really Associated with Increased Risk of Cancer? A Systematic Review and Meta-Analysis. Obes. Rev. 2021, 22, e13170. [Google Scholar] [CrossRef] [PubMed]
  127. Sharretts, J.; Galescu, O.; Gomatam, S.; Andraca-Carrera, E.; Hampp, C.; Yanoff, L. Cancer Risk Associated with Lorcaserin—The FDA’s Review of the CAMELLIA-TIMI 61 Trial. N. Engl. J. Med. 2020, 383, 1000–1002. [Google Scholar] [CrossRef]
  128. Jeffrey Conn, P.; Christopoulos, A.; Lindsley, C.W. Allosteric Modulators of GPCRs: A Novel Approach for the Treatment of CNS Disorders. Nat. Rev. Drug Discov. 2009, 8, 41–54. [Google Scholar] [CrossRef] [Green Version]
  129. García-Cárceles, J.; Decara, J.M.; Vázquez-Villa, H.; Rodríguez, R.; Codesido, E.; Cruces, J.; Brea, J.; Loza, M.I.; Alén, F.; Botta, J.; et al. A Positive Allosteric Modulator of the Serotonin 5-HT 2C Receptor for Obesity. J. Med. Chem. 2017, 60, 9575–9584. [Google Scholar] [CrossRef]
  130. Singh, K.; Sona, C.; Ojha, V.; Singh, M.; Mishra, A.; Kumar, A.; Siddiqi, M.I.; Tripathi, R.P.; Yadav, P.N. Identification of Dual Role of Piperazine-Linked Phenyl Cyclopropyl Methanone as Positive Allosteric Modulator of 5-HT2C and Negative Allosteric Modulator of 5-HT2B Receptors. Eur. J. Med. Chem. 2019, 164, 499–516. [Google Scholar] [CrossRef]
  131. Cifuentes, L.; Eckel-Passow, J.; Acosta, A. Precision Medicine for Obesity. Dig. Dis. Interv. 2021, 5, 239. [Google Scholar] [CrossRef] [PubMed]
  132. Severin, R.; Sabbahi, A.; Mahmoud, A.M.; Arena, R.; Phillips, S.A. Precision Medicine in Weight Loss and Healthy Living. Prog. Cardiovasc. Dis. 2019, 62, 15. [Google Scholar] [CrossRef] [PubMed]
  133. Hurtado, A.M.D.; Acosta, A. Precision Medicine and Obesity. Gastroenterol. Clin. N. Am. 2021, 50, 127. [Google Scholar] [CrossRef] [PubMed]
  134. Griebsch, N.I.; Kern, J.; Hansen, J.; Rullmann, M.; Luthardt, J.; Helfmeyer, S.; Dekorsy, F.J.; Soeder, M.; Hankir, M.K.; Zientek, F.; et al. Central Serotonin/Noradrenaline Transporter Availability and Treatment Success in Patients with Obesity. Brain Sci. 2022, 12, 1437. [Google Scholar] [CrossRef]
  135. He, Y.; Brouwers, B.; Liu, H.; Liu, H.; Lawler, K.; Mendes de Oliveira, E.; Lee, D.K.; Yang, Y.; Cox, A.R.; Keogh, J.M.; et al. Human Loss-of-Function Variants in the Serotonin 2C Receptor Associated with Obesity and Maladaptive Behavior. Nat. Med. 2022, 28, 2537. [Google Scholar] [CrossRef]
  136. Wold, E.A.; Garcia, E.J.; Wild, C.T.; Miszkiel, J.M.; Soto, C.A.; Chen, J.; Pazdrak, K.; Fox, R.G.; Anastasio, N.C.; Cunningham, K.A.; et al. Discovery of 4-Phenylpiperidine-2-Carboxamide Analogues as Serotonin 5-HT2C Receptor-Positive Allosteric Modulators with Enhanced Drug-like Properties. J. Med. Chem. 2020, 63, 7529. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of the 5-HT2CR function modulation by agonists and positive-allosteric modulators (PAMs). The 5-HT2CR agonists bind to the same orthosteric sites as endogenous serotonin (5-HT) at the 5-HT2CR to evoke a response. Opposed to the 5-HT2CR agonists, the 5-HT2CR PAMs bind to allosteric sites at the 5-HT2CR, enhancing the affinity and efficacy of the 5-HT2CR response by 5-HT.
Figure 1. Mechanisms of the 5-HT2CR function modulation by agonists and positive-allosteric modulators (PAMs). The 5-HT2CR agonists bind to the same orthosteric sites as endogenous serotonin (5-HT) at the 5-HT2CR to evoke a response. Opposed to the 5-HT2CR agonists, the 5-HT2CR PAMs bind to allosteric sites at the 5-HT2CR, enhancing the affinity and efficacy of the 5-HT2CR response by 5-HT.
Nutrients 15 01449 g001
Table 1. Description of antiobesity drugs affinity to 5-HT1-2 receptor (5-HT1-2R) subtypes, type of binding and mechanisms of 5-HT2C receptor (5-HT2CR) activation.
Table 1. Description of antiobesity drugs affinity to 5-HT1-2 receptor (5-HT1-2R) subtypes, type of binding and mechanisms of 5-HT2C receptor (5-HT2CR) activation.
DrugAffinity to 5-HT1-2R Subtypes *Selectivity to 5-HT2CRMechanism of 5-HT2CR Activation
piperazine
derivatives
(e.g., m-chlorophenyl piperazine)
5-HT2B ≥ 5-HT2C > 5-HT2A > 5-HT1A ≥ 5-HT1B [33]nonselective
ligand
indirect agonism
(affinity to 5-HT transporter and followed by 5-HT reuptake inhibition or 5-HT release enhancement)
fenfluramines5-HT2B ≥ 5-HT2C > 5-HT2A [34]nonselective
ligand
indirect agonism
(affinity to 5-HT transporter and followed by 5-HT reuptake inhibition)
sibutramine5-HT1A = 5-HT1B = 5-HT2A = 5-HT2C [35]nonselective
ligand
indirect agonism
(affinity to 5-HT transporter and followed by 5-HT reuptake inhibition)
lorcaserin5-HT2C > 5-HT2A > 5-HT2B [36]selective
ligand
direct agonism
* mostly important for food intake and unwanted effects.
Table 2. Efficacy of 5-HT2C receptor agonist properties in double-blind, placebo-controlled, randomized clinical obesity trials. The patient’s inclusion criteria are increased body mass index (BMI; >25 kg/m2) or ideal body weight (IBW; 120–180%). The mean weight loss (kg or %) of the drug group (D) is compared to the placebo group (P). Multicenter studies were marked by an asterisk (*). QD—once-a-day treatment; BID—two times-a-day treatment.
Table 2. Efficacy of 5-HT2C receptor agonist properties in double-blind, placebo-controlled, randomized clinical obesity trials. The patient’s inclusion criteria are increased body mass index (BMI; >25 kg/m2) or ideal body weight (IBW; 120–180%). The mean weight loss (kg or %) of the drug group (D) is compared to the placebo group (P). Multicenter studies were marked by an asterisk (*). QD—once-a-day treatment; BID—two times-a-day treatment.
DrugDosage (mg)Duration (Months)Patients Completed Study
D|P
Key Inclusion Criteria
(BMI > 25 kg/m2 or IBW 120–180%)
Mean Weight Loss
D vs. P (kg or %)
Ref.
d-fenfluramine10 BID385|85 *120–180%2.79 vs. 2.83 kg[70]
15 BID168|169120–180%5.84 vs. 1.85 kg[71]
12|1228–35 kg/m23.1 ± 2.3 vs. 0.1 ± 1.2 kg[72]
12404|418 *≥120%9.82 vs. 7.15 kg[73]
30 QD
or
BID
382|85 *120–180%5.63 vs. 2.83 kg[70]
30|30120–180%4.6 ± 1.6 kg vs. no changed[74]
1236|39≥135%12.8 vs. 8.6 kg[75]
60 BID387|85 *120–180%7.23 vs. 2.83 kg[70]
Sibutramine5 QD219|20130–180%2.9 ± 2.3 vs. 1.4 ± 2.1 kg[76]
356|59 *27–40 kg/m22.4 ± 0.5 vs. 1.4 ± 0.5 kg[77]
6107|87 *30–40 kg/m23.7 vs. 1.3 kg[78]
10 QD359|59 *27–40 kg/m25.1 ± 0.5 vs. 1.4 ± 0.5 kg[77]
699|87 *30–40 kg/m25.7 vs. 1.3 kg[78]
104|94 *≥30 and ≤40 kg/m28.2 vs. 3.9 kg[79]
402933|2825 *25–45 kg/m21.7 vs. +0.7 kg[80]
15 QD362|59 *27–40 kg/m22 4.9 ± 0.5  vs. 1.4 ± 0.5 kg[77]
698|87 *30–40 kg/m27.0 vs. 1.3 kg[78]
12281|80±44 kg/m26.5 ± 0.31 vs. 1.9 ± 0.56 kg[81]
68|64 *>27 kg/m25.5 ± 0.6 vs. 0.2 ± 0.5 kg[82]
~14114|103 *≥30 and <40 kg/m28.1 ± 8.2 vs. 5.1 ± 6.5 kg[83]
20 QD221|20130–180%5.0 ± 2.7 vs. 1.4 ± 2.1[76]
696|87 *30–40 kg/m28.2 vs. 1.3 kg[78]
151|152 *≥27 kg/m24.9 vs. 0.6 kg[84]
1262|64 *>27 kg/m28.0 ± 0.9 vs. 0.2 ± 0.5 kg[82]
30 QD6101|87 *30–40 kg/m29.0 vs. 1.3 kg[78]
Lorcaserin10 QD
or
10 BID
~229|2827–45 kg/m23.8 ± 0.4 vs. 2.2 ± 0.5 kg[85]
386 or 77|88 *30–45 kg/m21.8 or 3.6 vs. 0.3 kg[86]
659|53 *≥33 and ≤55 kg/m22.4 ± 0.8 vs. +0.6 ± 0.8 kg[87]
12748|243 *≥27 kg/m24.2 vs. 1.4 kg[88]
1275 or 169|157 *27–45 kg/m244.7 or 37.5 vs. 16.1%[89]
275|684 *27–45 kg/m25.81 ± 0.16 vs. 2.16 ± 0.14%[90]
131800|155030–45 kg/m247.1 vs. 22.6%[91]
24564|684 *27–45 kg/m27.0 ± 0.2 vs. 3.0 ± 0.2%[90]
40748|243 *≥27 kg/m24.0 kg vs. 2.1 kg[88]
15 QD382|88 *30–45 kg/m22.6 vs. 0.3 kg[86]
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

Przegaliński, E.; Witek, K.; Wydra, K.; Kotlińska, J.H.; Filip, M. 5-HT2C Receptor Stimulation in Obesity Treatment: Orthosteric Agonists vs. Allosteric Modulators. Nutrients 2023, 15, 1449. https://doi.org/10.3390/nu15061449

AMA Style

Przegaliński E, Witek K, Wydra K, Kotlińska JH, Filip M. 5-HT2C Receptor Stimulation in Obesity Treatment: Orthosteric Agonists vs. Allosteric Modulators. Nutrients. 2023; 15(6):1449. https://doi.org/10.3390/nu15061449

Chicago/Turabian Style

Przegaliński, Edmund, Kacper Witek, Karolina Wydra, Jolanta H. Kotlińska, and Małgorzata Filip. 2023. "5-HT2C Receptor Stimulation in Obesity Treatment: Orthosteric Agonists vs. Allosteric Modulators" Nutrients 15, no. 6: 1449. https://doi.org/10.3390/nu15061449

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