Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jul 1.
Published in final edited form as: Physiol Behav. 2024 Apr 23;281:114565. doi: 10.1016/j.physbeh.2024.114565

Targeting GLP-1 receptors to reduce nicotine use disorder: Preclinical and clinical evidence

Rae J Herman a, Heath D Schmidt b,c,*
PMCID: PMC11128349  NIHMSID: NIHMS1991125  PMID: 38663460

Abstract

Nicotine use disorder (NUD) remains a leading cause of preventable death in the U.S. Unfortunately, current FDA-approved pharmacotherapies for smoking cessation have limited efficacy and are associated with high rates of relapse. One major barrier to long-term smoking abstinence is body weight gain during withdrawal. Nicotine withdrawal-induced body weight gain can also lead to development of chronic disease states like obesity and type II diabetes mellitus. Therefore, it is critical to identify novel pharmacotherapies for NUD that decrease relapse and nicotine withdrawal symptoms including body weight gain. Recent studies demonstrate that glucagon-like peptide-1 receptor (GLP-1R) agonists attenuate voluntary nicotine taking and seeking and prevent withdrawal-induced hyperphagia and body weight gain. Emerging evidence also suggests that GLP-1R agonists improve cognitive deficits, as well as depressive- and anxiety-like behaviors, which contribute to smoking relapse during withdrawal. While further studies are necessary to fully characterize the effects of GLP-1R agonists on NUD and understand the mechanisms by which GLP-1R agonists decrease nicotine withdrawal-mediated behaviors, the current literature supports GLP-1R-based approaches to treating NUD.

Keywords: Smoking, Relapse, Self-administration, Withdrawal, Hyperphagia, Semaglutide

1. Introduction

Nicotine consumption, whether through tobacco smoking or electronic nicotine delivery systems (i.e., e-cigarettes), remains a global health concern [1]. In 2021, approximately 46 million (18.7 %) U.S. adults reported using nicotine products (11.5 % of U.S. adults reported smoking traditional cigarettes, while 4.5 % reported using e-cigarettes) [2]. The rising popularity of e-cigarettes has led to an increase in youth nicotine product use with 2.55 million middle and high school students reporting e-cigarette use in 2022 [3]. Of these students, four in ten reported frequent use, and one in four reported daily use of e-cigarettes. Furthermore, 76 % of middle and high school students who consume e-cigarettes reported using another nicotine product, and adolescents who use e-cigarettes are 3.5 times more likely to initiate traditional cigarette smoking than adolescents who don’t use e-cigarettes [46]. Smoking cessation at any age significantly increases life expectancy [7]. Approximately 68 % of adult cigarette smokers want to quit smoking, while 65.3 % of adolescent tobacco users seriously consider quitting all tobacco products [8,9]. Despite the well-known negative effects of nicotine and the large proportion of smokers that desire to quit smoking, fewer than one in ten smokers who attempt to quit are successful [8,10]. Moreover, 67.4 % of current e-cigarette users have tried unsuccessfully to quit vaping, in large part due to craving and withdrawal symptoms similar to traditional cigarette smokers [11,12]. Thus, given the health risks of vaping and the increased risk of tobacco smoking in a growing population of e-cigarette users, it is more important than ever to develop more effective treatments for nicotine use disorder (NUD).

Concerns about weight gain are a major barrier to achieving long-term smoking abstinence for both men and women [1315]. Smoking cessation increases consumption of highly palatable foods, and 80–90 % of smokers who quit gain weight [1621]. Most smokers who quit gain an average of 5–15 pounds in the first few months of abstinence, while some smokers (~14 %) gain more than 20 pounds [19]. Nicotine withdrawal-induced body weight gain contributes to an increased risk of type II diabetes mellitus (T2DM) and hypertension and can prevent cessation-induced improvements in lung function [19,2224]. These health hazards are amplified by the high comorbidity of smoking with metabolic and cardiovascular diseases [15,25,26]. Therefore, it is critical to consider nicotine withdrawal-induced body weight gain as a potential health risk during smoking cessation.

FDA-approved treatments for NUD, including nicotine replacement therapy (NRT), varenicline, and bupropion, decrease smoking relapse. However, their long-term efficacy is modest, and these treatments delay, but do not prevent, body weight gain during smoking abstinence [15,19, 2729]. Therefore, it is crucial to identify novel pharmacotherapies that promote long-term abstinence while minimizing withdrawal-induced hyperphagia and body weight gain [15,19]. Glucagon-like peptide 1 receptor (GLP-1R) agonists are FDA-approved for treating T2DM and obesity [3032]. Mounting evidence indicates that GLP-1R agonists reduce the rewarding and reinforcing effects of licit and illicit drugs including nicotine [3337]. Importantly, recent studies showed that GLP-1R agonist monotherapy attenuated nicotine withdrawal-induced hyperphagia and body weight gain in male and female rats [38]. GLP-1R agonists also decreased voluntary nicotine taking and seeking in rodent models [38,39], and may improve other nicotine withdrawal-related phenotypes including cognitive deficits and mood disorders [4042]. Together, these results suggest that GLP-1R agonists could be re-purposed for treating NUD, either as a monotherapy or as an adjunct therapy in combination with other NUD treatments. Here, we review the preclinical and clinical evidence supporting GLP-1R agonists as novel NUD pharmacotherapies.

2. GLP-1 and GLP-1Rs

GLP-1 is an incretin hormone and neuropeptide produced peripherally in L cells of the small intestine and centrally in the nucleus tractus solitarius (NTS) of the caudal brainstem [4345]. GLP-1Rs are G-protein coupled receptors expressed pre- and post-synaptically throughout the brain, including nuclei within the mesolimbic dopamine system [4648]. GLP-1Rs are predominantly Gs-coupled and activate cells via increased cAMP signaling [49]. However, GLP-1R activation also recruits β-arrestin to increase ERK1/2 signaling [50,51]. Differential activation of these downstream pathways, or biased agonism, likely occurs due to differences in ligand binding to extracellular loops on the GLP-1R [5153]. The relative contributions of intracellular calcium, cAMP signaling, and ERK1/2 signaling to the cellular and behavioral effects of GLP-1R agonists are not clear [54]. Since GLP-1R agonists like liraglutide, semaglutide, dulaglutide and exenatide activate both cAMP and β-arrestin pathways, future studies should consider the potential of biased agonism when developing and testing novel GLP-1R agonists [55].

3. GLP-1R agonists

Given the role of endogenous GLP-1 in regulating food intake and glucose homeostasis, GLP-1R agonists have been developed to treat T2DM and obesity [3032,56]. Endogenous GLP-1 has a very short half-life in human plasma of only 1–2 min due to rapid degradation by the enzyme dipeptidyl peptidase IV (DPP-IV) [47]. This pharmacokinetic profile limits the therapeutic potential of GLP-1. Serendipitously, Exendin-4 (trade name Byetta) was isolated from the venom of the lizard Heloderma suspectum (Gila monster) and found to function as an agonist of the GLP-1R [57]. Exendin-4 is 53 % homologous to human GLP-1, and resistant to DPP-IV degradation due to a serine-alanine amino acid substitution on the N-terminus [47,58]. As such, Exendin-4 has a half-life of ~2.5 h in humans and, therefore, requires twice daily dosing as a subcutaneous injection. However, it is also available in an extended-release formulation (trade name Bydureon) that is administered once weekly [59]. Subsequently, second-generation GLP-1R agonists with improved pharmacokinetics were developed. For example, the GLP-1R agonist liraglutide (trade names Victoza and Saxenda) was created by modifying the endogenous GLP-1 peptide sequence (97 % homologous) – specifically, inserting an aminoisobutyric acid into amino acid 8 of the N-terminus to increase resistance to DPP-IV degradation [6062]. Liraglutide also has an added spacer and C-16 fatty monoacid side chain to enhance albumin binding and decrease renal clearance. These modifications impart a half-life of ~13 h which is amenable to once daily subcutaneous injection [58]. Other second-generation GLP-1R agonists incorporate different strategies, like conjugation with the Fc fragment of IgG to create dulaglutide (trade name Trulicity; 90 % homologous to GLP-1). Dulaglutide has a half-life of ~90 h and therefore is dosed once weekly [6365]. More recently, the third generation GLP-1R agonist semaglutide (trade names Ozempic, Wegovy, and Rybelsus) was developed. Semaglutide is 94 % homologous to human GLP-1, but with three modifications - 2 amino acid substitutions: one at position 8 (alanine to α-aminoisobutyric acid) and one at position 34 (lysine to arginine), and acylation of the lysine residue in position 26 with a spacer and C-18 fatty diacid chain - that increase its half-life to ~165–184 h making it amenable for once-weekly injection [6668]. The fatty diacid and spacer modifications impart stronger binding to albumin, while the amino acid substitution at position 8 renders semaglutide more resistant to DPP-IV degradation [68]. Semaglutide is the first GLP-1R agonist available in a daily oral tablet formulation (trade name Rybelsus) [31]. Semaglutide also has greater free drug plasma concentrations and stronger affinity for the GLP-1R than older generation GLP-1R agonists, which may explain its greater magnitude of effect on weight loss (two and three times greater than liraglutide and exenatide, respectively) [69,70]. Given this increased efficacy, GLP-1R agonists have surged in popularity in recent years as treatments for T2DM and obesity. Newer generation drugs function as dual incretin receptor agonists. For example, tirzepatide (recently FDA-approved to treat T2DM, trade name Mounjaro, and chronic weight management, trade name Zepbound) is an analogue of gastric inhibitory polypeptide (GIP) that functions as a dual agonist of GIP receptors and GLP-1Rs. Tirzepatide produces a greater reduction in body weight compared to semaglutide, demonstrating that dual agonist “twincretin” approaches may provide even more efficacious pharmacotherapies for T2DM and obesity in the future [7173].

There are important differences to consider with respect to the efficacy and therapeutic responses of GLP-1R agonists [74]. GLP-1R agonists can be categorized as short-acting or long-acting based on their half-lives and formulations [63]. These categories have diverging therapeutic effects [63,75]. Short-acting GLP-1R agonists (e.g., exenatide) lower blood glucose levels via glucose-stimulated insulin secretion from pancreatic beta cells, reduced glucagon release, and inhibition of gastric emptying [63,76]. In contrast, long-acting GLP-1R agonists (e.g., dulaglutide, extended-release exenatide, and liraglutide) have stronger efficacy to decrease fasting blood glucose levels through not only stimulation of insulin secretion and reduction of glucagon levels, but also a greater suppression of food intake [63]. Furthermore, meta-analyses of randomized controlled trials demonstrated differences in clinical outcomes based on GLP-1R agonist formulation [77]. Once-weekly GLP-1Rs, like extended-release exenatide and dulaglutide, were more effective in controlling glycaemia, but equally effective in decreasing body weight compared to twice daily exenatide in participants with T2DM [77]. Liraglutide was more effective in both decreasing body weight and controlling glycemia compared to twice daily exenatide. Analyses including the third generation GLP-1R agonist semaglutide supported the finding that long-acting GLP-1R agonists have the greatest effect on weight loss [78]. This study also demonstrated that semaglutide was the most effective GLP-1R agonist to reduce blood glucose levels. GI-related adverse effects (e.g., nausea, diarrhea, vomiting and dyspepsia) are the most common adverse effects associated with GLP-1R agonists [79]. GI adverse effects are often dose-dependent and can be improved by slower titration regimens [79]. Importantly, once-weekly GLP-1R agonists decreased the incidence of minor hypoglycemia compared to twice daily exenatide and did not increase the incidence of adverse effects [77].

Given that the therapeutic effects of GLP-1R agonists are due, at least in part, to activation of neuronal GLP-1Rs [80,81], one must also consider CNS distribution and central effects when comparing GLP-1R agonists. Interestingly, semaglutide and liraglutide have different brain penetrance, central distribution, and transcriptomic changes after systemic administration, indicating that GLP-1R agonists have distinct interactions with the central nervous system [60,82]. Systemically administered, fluorescently labeled liraglutide and semaglutide both bound to proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) positive neurons in the arcuate nucleus of the hypothalamus [82]. However, semaglutide showed higher fluorescent signal intensity in the lateral septal nucleus, septofimbrial nucleus, medial mammillary nucleus, and more extensive lateral distribution in the arcuate nucleus, while liraglutide showed higher binding in the vascular organ of the lamina terminalis, supraoptic nucleus, subfornical organ and paraventricular nucleus of the hypothalamus [82]. Semaglutide, but not liraglutide, penetrated the posterior region of the arcuate nucleus, a nucleus known to regulate food intake through its projections to the parabrachial nucleus [82,83]. Semaglutide and liraglutide produced unique brain region-specific transcriptomic changes following systemic administration [82]. For example, semaglutide penetrated more effectively into the arcuate nucleus and lateral septal nucleus, and produced greater differential expression of genes in these nuclei including genes regulating metabolic pathways, ribosomes, and oxidative phosphorylation [82]. These differences in brain penetrance and distribution may be important factors underlying the differences in efficacy between GLP-1R agonists. It is important to note that the extent of GLP-1R agonist penetrance across the blood brain barrier and the necessity of central GLP-1R activation for the therapeutic effects of GLP-1R agonists is under some debate, and may depend on the agonist studied or experimental timeframe [81,84]. GLP-1R agonists may use specialized mechanisms to access circum-ventricular organs as well as more distal brain regions through the blood-cerebrospinal fluid barrier [85]. Interestingly, administration of GLP-1R agonists is associated with activation of neurons in the brain that do not express GLP-1Rs [86], further underscoring the complexity of the mechanisms underlying the efficacy of GLP-1R agonists. Understanding the central mechanisms underlying the effects of GLP-1R agonists will inform the development of more targeted and effective treatments tailored for specific patient needs [63].

In Section 4 below, we will discuss preclinical and clinical findings related to GLP-1R agonists and nicotine-mediated behaviors. In Section 5, we will review preclinical and clinical studies related to GLP-1R agonists and nicotine withdrawal phenotypes. Ongoing clinical trials investigating the efficacy of GLP-1R agonists to treat NUD will be discussed in Section 6.

4. GLP-1R agonists and nicotine-mediated behaviors

4.1. GLP-1R agonists and nicotine reinforcement

Recent preclinical studies indicated that GLP-1R agonists decreased the rewarding and reinforcing effects of nicotine in rodents (Table 1). Systemic administration of Exendin-4 (2.4 μg/kg, i.p.) attenuated the expression of nicotine-induced conditioned place preference (CPP) in male mice, suggesting that GLP-1R agonists reduce the rewarding effects of nicotine [87]. Consistent with these effects, systemic administration of Exendin-4 (10 μg/kg, i.p.) or the DPP-IV inhibitor sitagliptin (10 mg/kg, i.p.) attenuated nicotine self-administration in male mice [39]. A recent study found that systemic administration of liraglutide (25 μg/kg, i.p) attenuated nicotine self-administration in male and female rats [38]. This study was the first to demonstrate a suppressive effect of a GLP-1R agonist on nicotine self-administration in females, which is particularly encouraging given that women often have greater difficulty quitting smoking and show poorer outcomes with NRT [88,89]. Together, these preclinical studies suggest that GLP-1R agonists attenuate the reinforcing efficacy of nicotine.

Table 1.

Effects of GLP-1R agonists on nicotine-mediated behaviors and neurochemical responses.

Study Sex/strain/species Model GLP-1R agonist pretreatment Route of admin Results
Egecioglu et al. [87] Male NMRI mice Nicotine-induced locomotor activation Acute Exendin-4 (0 or 2.4 μg/kg) i.p. Attenuated nicotine-induced locomotor activation
Nicotine-evoked dopamine release Acute Exendin-4 (0 or 2.4 μg/kg) i.p. Attenuated nicotine-evoked dopamine release in the NAc
Expression of nicotine CPP Acute Exendin-4 (0 or 2.4 μg/kg) i.p. Blocked expression of CPP
Expression of nicotine-induced locomotor sensitization Acute Exendin-4 (0 or 2.4 μg/kg) i.p. Attenuated expression of locomotor sensitization
Tuesta et al. [39] Male C57BL6/J mice Nicotine IVSA (0.1 mg/kg/infusion) Acute Exendin-4 (0 or 10 μg/kg) & sitagliptin (0 or 10 mg/kg) i.p. Attenuated nicotine self-administration
Male Wistar rats Nicotine IVSA (0.03 mg/kg infusion) Acute Exendin-4 (0 or 0.1 μg/0.5 μl/hemisphere) intra-IPN Attenuated nicotine self-administration
Change in ICSS threshold after nicotine IVSA Acute Exendin-4 (0 or 0.1 μg/0.5 μl/hemisphere) intra-IPN Prevented nicotine-induced changes in ICSS thresholds
Herman et al. [38] Male and female Sprague Dawley rats Nicotine IVSA (0.03 mg/kg/infusion) Acute liraglutide (0 or 25 μg/kg) i.p. Attenuated nicotine self-administration
Extinction of nicotine taking Repeated liraglutide (0 or 25 μg/kg) i.p. Decreased days to extinguish
Drug + cue-induced reinstatement of nicotine seeking Repeated liraglutide (0 or 25 μg/kg) i.p. Attenuated nicotine seeking during withdrawal
Falk et al. [95] Male NMRI mice Nicotine-evoked dopamine release Acute Liraglutide (0 or 10 nmol/kg) s.c. Attenuated nicotine-evoked dopamine release in the NAc

i.p. = intraperitoneal; CPP = conditioned place preference; IVSA = intravenous self-administration; ICSS = intra-cranial self-stimulation; IPN = interpeduncular nucleus; s.c. = subcutaneous; NAc = nucleus accumbens.

The neural mechanisms underlying the suppressive effects of GLP-1R agonists on nicotine taking are likely to be complex and involve neural circuits that mediate reward and aversion. Nicotine activated neurons in the interpeduncular nucleus (IPN) and constitutive GLP-1R knockout blocked these effects, suggesting that GLP-1R activation plays a role in the response of IPN neurons to nicotine [39]. The medial habenula (MHb) projects almost exclusively to the IPN, and this circuit regulates the aversive effects of higher doses of nicotine [9094]. Based on this anatomy, GLP-1R activation in MHb->IPN circuits may increase the aversive effects of nicotine to limit nicotine taking. GLP-1Rs are expressed on MHb afferents in the IPN, and pharmacological activation of these receptors attenuated nicotine self-administration [39]. These results indicate that Exendin-4 activates presynaptic GLP-1Rs on MHb terminals in the IPN to decrease nicotine taking. Consistent with these effects, knockdown of GLP-1Rs in the MHb or pharmacological inhibition of GLP-1Rs in the IPN increased nicotine taking [39]. Interestingly, GLP-1R knockdown in the MHb increased nicotine self-administration only on the descending limb of the dose-response curve [39], further supporting the hypothesis that GLP-1R activation in the MHb augments the aversive effects of higher unit doses of nicotine to decrease drug taking. While it is clear that activation of GLP-1Rs in the IPN decreased nicotine taking [39], future studies are needed to confirm whether activation of GLP-1Rs in the MHb->IPN circuit increases the aversive effects of nicotine, decreases the reinforcing efficacy of nicotine, or both.

GLP-1Rs are expressed throughout the brain, including the mesolimbic dopamine system [48]. Given that GLP-1R agonists decreased nicotine-evoked dopamine release in the NAc, it is possible and likely that GLP-1R agonism attenuates nicotine taking via effects on striatal dopamine signaling [87,95]. This hypothesis is supported by studies showing that activation of GLP-1Rs in the NAc or VTA was sufficient to reduce voluntary cocaine taking [9698]. Moreover, central GLP-1R activation reduced cocaine-evoked phasic dopamine signaling [99]. These findings suggest that GLP-1R agonists reduce nicotine taking, in part, by attenuating nicotine-evoked activation of the mesoaccumbens pathway. However, this question has not been investigated directly in rodents self-administering nicotine. Future work should determine the functional significance of NAc and VTA GLP-1Rs in nicotine taking to determine the relative contributions of the mesolimbic reward system versus MHb->IPN aversion circuits in the behavioral responses to GLP-1R agonists. Since systemically administered GLP-1R agonists activate neurons throughout the brain [100], it is likely that GLP-1R agonists influence activity of circuits involved in reward and aversion to reduce nicotine taking. Given that the patterns of distribution of GLP-1R agonists differ across the CNS, it is important to consider that individual GLP-1R agonists may differentially engage different neural circuits which may influence their efficacy in reducing nicotine taking and seeking [82].

4.2. GLP-1R agonists and extinction of nicotine-taking behavior

While there is not an extensive literature exploring the effects of GLP-1R agonism on extinction of drug taking, a recent study showed that repeated liraglutide administration during abstinence decreased the number of days required to extinguish nicotine self-administration [38]. In these tests, liraglutide was administered 10 min before each extinction training session [38]. These results are consistent with previous studies which showed that systemic administration of Exendin-4 facilitated extinction of cocaine-induced CPP and decreased subsequent cocaine priming-induced reinstatement of CPP in mice [101]. These findings suggest that the combination of GLP-1R agonist treatment and daily extinction training may facilitate extinction learning-related plasticity [38]. GLP-1Rs are expressed in nuclei known to mediate extinction of drug-taking behavior including the NAc shell and hypothalamic subregions, further supporting a potential role of central GLP-1Rs in extinction learning [48,102]. GLP-1R activation increased glutamate signaling and AMPA receptor trafficking in drug-naïve rats [103106]. Since enhanced glutamate signaling and upregulation of AMPA receptors facilitated extinction and decreased drug seeking [107, 108], GLP-1R agonists may enhance glutamate-mediated synaptic plasticity and facilitate extinction learning to reduce nicotine seeking during abstinence. To identify the mechanisms underlying the effects of GLP-1R agonists on extinction learning, future studies should investigate whether activation of GLP-1Rs in nuclei like the NAc shell and hypothalamus facilitate extinction of nicotine-taking behavior and decrease nicotine reinstatement. Studies should also investigate whether systemic GLP-1R activation increases glutamate signaling in the NAc shell and hypothalamus of nicotine-experienced rats and, if so, whether this increase is necessary for the effects of liraglutide on extinction rates during nicotine abstinence [38]. These experiments would expand our understanding of the role of central GLP-1Rs in extinction learning and the potential mechanisms underlying the suppressive effects of GLP-1R agonists on nicotine-seeking behaviors.

4.3. GLP-1R agonists and the reinstatement of nicotine-seeking behavior

In addition to reducing voluntary nicotine taking, novel treatments for NUD should also prevent smoking relapse and promote long-term abstinence. Smoking relapse is modeled in laboratory animals using the nicotine self-administration/extinction/reinstatement paradigm. This model has good predictive validity as an in vivo medication screen for NUD pharmacotherapies [109111]. For example, the FDA-approved smoking cessation pharmacotherapy varenicline attenuated both the reinstatement of nicotine-seeking behavior in rats and smoking relapse in humans [111113]. With regard to GLP-1R agonists, a recent study showed that repeated liraglutide administration during abstinence attenuated drug seeking elicited by a priming injection of nicotine and re-exposure to conditioned light cues (i.e., lights that were previously paired with nicotine infusions during the self-administration phase of the experiment) [38]. These results suggest that GLP-1R agonist monotherapy may be effective in preventing smoking relapse. Given that GLP-1R agonists decreased neural responses to reward-related cues in humans [114,115], it will be important to determine if GLP-1R agonists reduce the incentive salience of nicotine cues, the reinforcing efficacy of nicotine, or both during withdrawal.

Little is known about the neural mechanisms underlying the efficacy of GLP-1R agonists to attenuate the reinstatement of nicotine-seeking behavior. GLP-1R agonists attenuated nicotine-evoked dopamine release in the NAc and phasic dopamine responses to food predictive cues [87,95,116]. Therefore, it is possible that GLP-1R agonists decrease nicotine- and/or cue-induced phasic dopamine signaling to attenuate the reinstatement of nicotine-seeking behavior. This hypothesis is supported by studies demonstrating that pharmacological inhibition of dopamine receptors decreased nicotine- and cue-induced reinstatement [110,117]. Future studies should measure dopamine levels during nicotine reinstatement tests in animals pretreated with a GLP-1R agonist to determine if the efficacy of GLP-1R agonists to reduce nicotine seeking is associated with reduced NAc dopamine signaling. In addition, there is some evidence that nicotine seeking is associated with changes in glutamate transmission in the NAc [110]. Cue-induced nicotine seeking was associated with increased extracellular glutamate, spine head diameter, and AMPA/NMDA ratios in the NAc, suggesting that glutamatergic plasticity may be necessary for cue-induced nicotine seeking [110,118]. Therefore, pharmacotherapies that increase glutamatergic plasticity and transmission in the NAc during withdrawal may attenuate the reinstatement of nicotine seeking [110]. Given that GLP-1R activation increased glutamate signaling and AMPA receptor trafficking in drug-naïve rats [103106], future studies should investigate whether increased glutamatergic signaling underlies the efficacy of GLP-1R agonists to prevent the reinstatement of nicotine-seeking behavior. Changes in hippocampal long term-potentiation (LTP) may also promote nicotine seeking [110]. Chronic nicotine self-administration induced hippocampal LTP [119], which may facilitate learned associations between nicotine and environmental cues [110]. Nicotine-induced hippocampal LTP was associated with decreased activity of GABAergic interneurons [120] and GLP-1R agonism enhanced GABA release in the hippocampus [121]. Therefore, it is provocative to think that GLP-1R agonism may activate inhibitory hippocampal circuits to prevent LTP and decrease the incentive salience of nicotine-associated cues during withdrawal. Future studies should investigate changes in hippocampal GABA signaling and LTP during nicotine reinstatement tests in rats treated with a GLP-1R agonist. Altogether, these studies would expand our understanding of the potential mechanisms underlying the effects of GLP-1R agonists on reward-related cues.

4.4. Adverse effects of GLP-1R agonists in preclinical models of NUD

The translational potential of GLP-1R agonists for NUD may be limited by their propensity to elicit nausea and vomiting [122]. While these adverse effects can limit patient compliance, they are often transient and mainly occur during treatment initiation and/or after dose increases [74,123,124]. The incidence of adverse effects can be minimized by using an escalation protocol to induce tolerance before introducing higher doses [74]. To screen for these potential adverse effects, preclinical studies measure pica, a model of nausea-like behavior in which rats consume a non-nutritive substance, such as kaolin clay, in response to an emetic agent [125]. The behaviorally relevant dose of liraglutide (25 μg/kg) that reduced nicotine taking and seeking did not produce pica in male and female nicotine-experienced rats [38]. This liraglutide dose is lower than doses shown to suppress locomotor activity and produce malaise-like effects in drug-naïve rats, indicating a behaviorally selective effect on nicotine taking and seeking [38,126, 127]. Consistent with these findings, doses of Exendin-4 that reduced nicotine taking had no effect on food self-administration [39]. Thus, preclinical studies show that GLP-1R agonists reduced voluntary drug taking and seeking at doses that are well-tolerated in nicotine-experienced rats.

4.5. Limitations of current preclinical studies of GLP-1R agonists in models of NUD

While emerging preclinical studies support re-purposing GLP-1R agonists for treating NUD, there are several limitations to consider when analyzing the current literature. To date, no studies have investigated how GLP-1R agonist pretreatment shifts the nicotine self-administration dose-response curve. These studies are critical towards understanding how GLP-1R agonists alter the reinforcing efficacy of nicotine. Moreover, previous nicotine self-administration and reinstatement studies only examined the efficacy of one dose of a GLP-1R agonist (25 μg/kg liraglutide or 10 μg/kg Ex-4) [38,39]. More comprehensive dose-response curves for each GLP-1R agonist are needed to fully understand the behavioral and physiological effects of GLP-1R agonists in nicotine-experienced rodents. Another significant limitation of the current literature is the lack of a reference compound (i.e., FDA-approved NUD medications including varenicline). Future studies should aim to compare the efficacy of GLP-1R agonists directly to an established NUD treatment to clarify their effect size and potential as a monotherapy. Finally, future studies should also compare the efficacy of GLP-1R agonist monotherapy to a combinatorial treatment (i.e., GLP-1R agonist + varenicline or GLP-1R agonist + NRT) to identify the most efficacious pharmacotherapeutic approach to treating NUD.

4.6. Clinical studies of GLP-1R agonists and NUD

Findings from preclinical studies of GLP-1R agonists and nicotine taking/seeking have recently been translated into clinical trials. One double-blind, randomized, placebo-controlled trial investigated the effects of extended-release exenatide (2 mg weekly, s.c.) on nicotine abstinence, craving, withdrawal, and post-cessation body weight gain [128]. This study measured outcomes after six weeks of treatment in overweight and/or prediabetic participants. In addition to exenatide or placebo, all participants received daily 21 mg NRT patches and brief weekly smoking cessation counseling (10–20 min weekly). The study enrolled 84 participants (41 in the exenatide + NRT group and 41 in the placebo + NRT group). Participants who received exenatide + NRT had increased seven-day point prevalence abstinence (defined as self-report of no smoking in the past seven days and expired CO level ≤5 ppm) compared to participants who received placebo + NRT at week six. Specifically, after six weeks of treatment 46.3 % of participants receiving exenatide + NRT were abstinent compared to 26.8 % of participants treated with placebo + NRT. Participants in the exenatide + NRT group also had decreased craving for cigarettes. During smoking abstinence, exenatide + NRT decreased withdrawal symptoms compared to placebo + NRT. However, there were no effects of exenatide + NRT on withdrawal symptoms in participants that did not reach abstinence. As discussed in more detail in Section 5.2.2 below, exenatide + NRT also reduced post-cessation body weight gain after six weeks of treatment [128]. While these results were an encouraging demonstration of the efficacy of GLP-1R agonists to decrease smoking and cigarette craving in the clinic, a different trial found no differences in smoking abstinence or craving in participants treated with dulaglutide + varenicline versus placebo + varenicline [129]. In this study, 255 daily smokers (mean body weight = 176.6 lbs) were treated with 0.75 mg dulaglutide (n = 127) or placebo (n = 128) for one week. Participants were then treated with 1.5 mg dulaglutide or placebo for the next 11 weeks (for a total of 12 weeks of dulaglutide treatment). All participants also received behavioral counseling and varenicline pharmacotherapy. After 12 weeks of treatment, there were no effects of dulaglutide + varenicline on abstinence – defined by self-reported seven days smoking abstinence and expired CO level ≤10 ppm – compared to placebo + varenicline. There were also no effects of dulaglutide + varenicline on craving for smoking. However, dulaglutide + varenicline was effective in decreasing post-cessation body weight gain and improving glucose metabolism compared to placebo + varenicline. A questionnaire measuring withdrawal symptoms was not utilized in this study.

There are several potential reasons for the differential effects observed in these two studies. In Lengsfeld et al. [129], seven-day abstinence rates were much higher than anticipated (63 % with dulaglutide + varenicline and 65 % with placebo + varenicline). Increased abstinence rates may have been due to frequent contact with study staff and feedback on carbon monoxide measurements used to measure nicotine exposure [129]. In contrast, Yammine et al. [128] found that 46.3 % of liraglutide + NRT-treated participants were abstinent for at least seven days following six weeks of treatment, compared to 26.8 % of vehicle + NRT-treated participants. These results suggest that high quit rates may have concealed any potential effects of dulaglutide on nicotine abstinence [129]. There are other key differences that should be considered when comparing these studies. Yammine et al. [128] utilized NRT as an adjunct therapy instead of varenicline, and only participants with prediabetes or that were overweight were included. Furthermore, the study population was predominantly older, black, and male. In contrast, Lengsfeld et al. [129] did not specify that participants must have prediabetes or be overweight, and their study group was predominantly younger, female, and white. Differences in GLP-1R agonists should also be considered when comparing these results, as pharmacokinetic and pharmacodynamic profiles differ between GLP-1R agonists (as discussed above) and may therefore affect therapeutic outcomes [63, 75].

4.7. Adverse effects of GLP-1R agonists in clinical studies of NUD

Adverse effects in Yammine et al. [128] were reported in four (9.5 %) participants in the exenatide + NRT group and one (2.3 %) participant in the placebo + NRT group. There was no significant difference in the percentage of adverse effects between treatment groups. In the exenatide + NRT group, adverse effects were injection site nodules that were mild in severity, resolved within two weeks, and did not result in treatment discontinuation. There were no reports of hypoglycemia, nausea, vomiting, dyspepsia, diarrhea, constipation, or tachycardia in either group. In contrast, Lengsfeld et al. [129] found higher rates of adverse effects during GLP-1R agonist treatment. GI distress was the most notable adverse effect (90 % of participants treated with dulaglutide + varenicline and 81 % of participants treated with placebo + varenicline reported GI distress at any time during treatment). Dulaglutide + varenicline-treated participants had more severe GI symptoms that required treatment (e.g., proton-pump inhibitors, antiemetics) more often. Specifically, 28 (22 %) participants in the dulaglutide + varenicline-treated group required treatment versus 14 (11 %) in the placebo + varenicline group. Participants in the dulaglutide + varenicline group were 2.5 times as likely as the placebo group to experience nausea. However, few participants withdrew due to adverse effects (dulaglutide + varenicline, n = 10; placebo + varenicline, n = 6). Differences in adverse effects between the studies by Lengsfeld et al. [129] and Yammine et al. [128] could be due to co-treatment with varenicline vs. NRT and/or differences in participant demographics as discussed above. Importantly, a recent meta-analysis of GLP-1R agonist treatment in overweight or obese patients without diabetes found no differences in adverse GI effects between participants treated with semaglutide and placebo [130]. However, exenatide and liraglutide both produced adverse GI effects greater than placebo [130]. These results suggest that newer generation GLP-1R agonists may have more favorable adverse effect profiles. Future research should compare the frequency and severity of adverse effects across studies with different adjunct treatments, patient populations, and GLP-1R agonists to identify treatment protocols that minimize the incidence of adverse effects and maximize patient compliance and treatment efficacy.

5. GLP-1R agonists and nicotine withdrawal symptoms

5.1. Nicotine withdrawal symptoms

The Diagnostic and Statistical Manual of Mental Disorders (DSM-5) describes seven main symptoms of nicotine withdrawal: irritability/anger/frustration, anxiety, difficulty concentrating, increased appetite, restlessness, depressed mood, and insomnia. For simplicity, these symptoms can be categorized as affective, somatic, or cognitive [131]. Affective symptoms include anxiety, anhedonia, depression, dysphoria, hyperalgesia, and irritability. Somatic symptoms include tremors, bradycardia, gastrointestinal discomfort, and increased appetite. Cognitive symptoms include difficulty concentrating and impaired memory [132]. Concerns about experiencing withdrawal symptoms are associated with decreased intention to quit [133] and increased smoking relapse [134]. Indeed, higher negative affect, craving, and composite withdrawal symptoms increase the likelihood of subsequent smoking relapse [135]. Furthermore, there is a cyclical relationship between withdrawal symptoms and relapse (i.e., relapse to smoking leads to subsequent increases in withdrawal symptoms such as negative affect), [135]. This cycle makes it even more important to address withdrawal symptoms in order to sustain long-term smoking abstinence. Nicotine withdrawal symptoms can be measured in rodent models of NUD after abrupt cessation of nicotine exposure or administration of a nicotinic acetylcholine receptor antagonist to precipitate withdrawal [136,137]. As in humans, withdrawal symptoms are alleviated by nicotine administration (e.g., NRT) or administration of a pharmacotherapy such as varenicline or bupropion [138]. Therefore, rodent models can be used to identify the neural substrates underlying nicotine withdrawal phenotypes and test the efficacy of potential treatments to ameliorate nicotine withdrawal symptoms.

5.2. GLP-1R agonists and nicotine withdrawal-induced hyperphagia and body weight gain

Nicotine consumption directly influences appetite and body weight. Adult smokers weigh less than non-smokers and are less likely to be overweight or obese [139]. Smokers tend to gain weight after quitting, and nicotine withdrawal-induced body weight gain is highly variable [18,139]. Smoking cessation is associated with an average weight gain of seven to 19 pounds within eight years of quitting [140,141]. In contrast, participants who continue to smoke gain, on average, 4–5 pounds of body weight [140]. As many as 13 % of those who quit smoking gain at least 23 pounds [18]. Body weight gain is greatest in the first two months of abstinence, but body weight can continue to increase for six months or more thereafter [15,142,143]. Concerns about post-cessation body weight gain are a major barrier to achieving long-term smoking abstinence for both men and women [1315,144]. Furthermore, nicotine withdrawal-induced body weight gain contributes to an increased risk of T2DM and hypertension [15,19,22,24]. Given the prevalence of nicotine withdrawal-induced body weight gain and its role as a major barrier to maintaining long-term smoking abstinence, it is critical to develop pharmacotherapies for NUD that prevent or reduce body weight gain during withdrawal, particularly in patient populations at a higher risk of excessive body weight gain. In this section, we will review preclinical and clinical evidence supporting GLP-1R agonists as potential treatments for nicotine withdrawal-induced body weight gain.

5.2.1. Preclinical studies of GLP-1R agonists and nicotine withdrawal-induced hyperphagia and body weight gain

Nicotine withdrawal is associated with increased consumption of high sugar and high fat food [21,145]. Therefore, novel medications that reduce nicotine withdrawal-induced hyperphagia and body weight gain should reduce consumption of highly palatable food. Two recent pre-clinical studies that support re-purposing GLP-1R agonists as treatments for nicotine withdrawal-induced body weight gain utilized high fat diet (HFD) in their models. In the first study, male mice maintained on HFD were implanted with subcutaneous osmotic minipumps that released nicotine at a rate of 15 mg/kg/day [146]. After 14 days of exposure, the minipumps were surgically removed and replaced with new minipumps containing nicotine, vehicle, or Exendin-4 (2.5 or 5.0 μg/kg/day). Body weight and energy consumption were measured for 14 days after minipump replacement. Nicotine-vehicle (N–V) mice gained significantly more weight than nicotine-nicotine (N–N) mice, with the highest incremental change in body weight during the first three days after minipump replacement. N–V mice continued to consume more food than N–N mice for one week after minipump replacement. In one experiment that utilized a pair-fed design, N–V mice were maintained on a HFD amount equal to that consumed by N–N mice. Pair-fed N–V mice did not demonstrate body weight gain after nicotine cessation, indicating that increased food intake is critical for nicotine withdrawal-induced body weight gain [146]. Chronic exposure to both doses of Exendin-4 during nicotine withdrawal significantly decreased both food intake and body weight gain compared to N–V rats [146]. Together, these preclinical findings suggest that GLP-1R agonists may attenuate nicotine withdrawal-induced hyperphagia and body weight gain in humans with NUD.

One potential limitation of this study is that continuous nicotine exposure via osmotic minipumps does not model the pattern of nicotine intake in human smokers, who voluntarily consume nicotine throughout the day. To more accurately model nicotine consumption patterns in humans, a recent study allowed male and female rats to self-administer nicotine (0.03 mg/kg/inf) during twice daily two-hour operant sessions over the course of 22 days [38]. Each rat that was allowed to respond for contingent infusions of nicotine was paired with a yoked rat that received infusions of saline. While lever pressing for the yoked saline rats had no scheduled consequences, these rats received the same number and temporal pattern of infusions as self-administered by their paired nicotine-experimental rat. Liraglutide (25 μg/kg, i.p.) or vehicle was administered daily beginning on the last day of self-administration and continued throughout 10 days of subsequent abstinence. All rats had ad libitum access to HFD during the self-administration and abstinence phases. Male and female rats experiencing nicotine withdrawal (nicotine-experimental group) consumed more HFD and gained more body weight than yoked-saline controls. Importantly, both hyperphagia and body weight were significantly reduced in nicotine-experienced rats treated with liraglutide compared to nicotine-experienced rats treated with vehicle. This study was the first to demonstrate that GLP-1R agonist monotherapy is sufficient to decrease hyperphagia and body weight gain during nicotine withdrawal.

Together, the studies by Takeda et al. [146] and Herman et al. [38] provide strong support for re-purposing GLP-1R agonists to reduce or prevent nicotine withdrawal-induced hyperphagia and body weight gain. Notably, both studies utilized HFD instead of normal chow. There is some evidence that nicotine withdrawal-induced hyperphagia may not occur in rodents given access to bland or normal chow [38, 146150]. Therefore, it is important to consider chow palatability as a potential factor modulating nicotine withdrawal-induced hyperphagia. Nonetheless, by demonstrating that GLP-1R agonists attenuate consumption of highly palatable food during nicotine withdrawal, these studies address an important question of high clinical relevance.

5.2.2. Clinical studies of GLP-1R agonists and nicotine withdrawal-induced hyperphagia and body weight gain

Consistent with the aforementioned preclinical studies, recent clinical trials showed that GLP-1R agonists reduced nicotine withdrawal-induced hyperphagia and body weight gain (Table 2). In one pilot, randomized, controlled trial (described above in Section 4.6), prediabetic and/or overweight smokers received once-weekly placebo (n = 41) or extended-release exenatide (2 mg, s.c.) (n = 41), along with NRT (21 mg patches) and brief smoking cessation counseling (10–20 min weekly) [128]. After six weeks of treatment, body weight was 5.6 pounds lower for the exenatide + NRT-treated group compared to placebo + NRT-treated controls. Participants treated with exenatide + NRT lost an average of 0.3 % of their baseline body weight over the course of treatment, while participants treated with placebo + NRT gained an average of 1.4 % of their baseline body weight. In a second study (described above in Section 4.6), treatment-seeking smokers were recruited for a randomized, double-blind, placebo-controlled, parallel group trial [129]. All participants were treated with the partial nicotinic receptor agonist varenicline along with subcutaneous injections of either dulaglutide (n = 127) (0.75 mg/0.5 ml in week 1, 1.5 mg/0.5 ml in week 2–12) or vehicle (n = 128). Participants also underwent behavioral counseling. Dulaglutide + varenicline treatment decreased withdrawal-induced body weight gain compared to placebo + varenicline after 12 weeks of treatment. Specifically, participants treated with dulaglutide + varenicline lost an average of 1.3 % of their baseline body weight, while participants treated with placebo + varenicline gained an average of 2.3 % of their baseline body weight. Additionally, dulaglutide + varenicline treatment decreased median HbA1c levels - an average of blood sugar level over the past 90 days and an indicator of prediabetes or T2DM - compared to placebo + varenicline. HbA1c decreased from 5.3 to 5.1 for participants treated with dulaglutide + varenicline. In contrast, mean HbA1c remained the same before and after treatment (5.4) in participants treated with placebo + varenicline [151]. New onset prediabetes (HbA1c between 5.7 % and 6.4 %) was more frequent in participants treated with placebo + varenicline than participants treated with dulaglutide + varenicline during smoking abstinence. Therefore, GLP-1R agonists may help to prevent or delay the development of diabetes in abstinent smokers. A 12-month follow-up of this study found that the beneficial effects of dulaglutide + varenicline treatment on body weight and HbA1c compared to varenicline alone gradually subsided after treatment discontinuation [152], emphasizing the importance of continued GLP-1R agonist treatment to maintain positive effects on body weight and blood glucose. These findings are consistent with previous studies demonstrating weight regain after cessation of GLP-1R treatment [153,154].

Table 2.

Clinical studies of GLP-1R agonists and nicotine use disorder.

Publication/ClinicalTrials.gov ID Clinical population Outcome measures GLP-1R agonist Study length Results/Study status
Lengsfeld et al. [129] 255 smokers 18–75 y/o with at least moderate nicotine dependence
(n = 128 placebo + varenicline, n = 127 dulaglutide + varenicline)
Smoking abstinence Craving for smoking Body weight change HbA1c levels Dulaglutide (1.5 mg weekly) or placebo in addition to smoking cessation counseling and oral varenicline (2 mg/daily) 12 weeks No effect on abstinence or craving Reduced post-cessation body weight gain Decreased HbA1c levels
Yammine et al. [128] 84 treatment-seeking smokers 18–75 y/o with prediabetes and/or overweight
(n = 41 placebo + NRT, n = 41 exenatide + NRT)
Smoking abstinence Craving for smoking Self-reported withdrawal Body weight change Extended-release exenatide (2 mg weekly) or placebo in addition to smoking cessation counseling and NRT (21 mg/daily) 6 weeks Increased abstinence Decreased craving Decreased withdrawal Reduced post-cessation body weight gain
NCT03712098 40 treatment-seeking smokers 18 y/o and older with overweight and obesity Smoking abstinence Body weight change Calories per day Liraglutide (escalating doses of 0.6–3.0 mg weekly) or placebo in addition to smoking cessation counseling 32 weeks Study completed, results not yet published
NCT05530577 Treatment-seeking smokers 21–65 y/o with at least moderate nicotine dependence Nicotine self-administration Reinstatement of nicotine seeking Daily cigarette smoking Craving for and subjective responses to smoking Body weight change HbA1c levels Semaglutide (escalating doses of 0.25–1.0 mg weekly) 10 weeks Currently recruiting
NCT05610800 Treatment-seeking smokers 18–75 y/o with prediabetes and/or overweight Smoking abstinence Body weight change Neural responses to visual stimuli Extended-release exenatide (2 mg weekly) or placebo in addition to smoking cessation counseling and NRT (de-escalating doses of 21–7 mg/daily) 14 weeks Currently recruiting
NCT06173778 Treatment-seeking smokers 18–75 y/o with prediabetes and/or overweight Body weight change Body fat mass change Waist circumference change Semaglutide (escalating doses of 0.24–2.4 mg weekly) in addition to smoking cessation counseling and NRT (de-escalating doses of 21–7 mg/daily) 28 weeks Not yet recruiting

y/o = years old; NRT = nicotine replacement therapy.

As discussed above, Yammine et al. [128] found no significant difference in the percentage of adverse effects between treatment groups. Adverse effects in the exenatide + NRT group were mild and transient. Moreover, there was no hypoglycemia, nausea, vomiting, dyspepsia, diarrhea, constipation, or tachycardia in either group. However, Lengsfeld et al. [129] found that participants treated with dulaglutide + varenicline had more severe GI symptoms that required treatment and were 2.5 times more likely to experience nausea than placebo + varenicline-treated participants. Nonetheless, few participants withdrew due to adverse effects (dulaglutide n = 10; placebo n = 6). Differences in the incidence of adverse effects may be due to co-treatment with varenecline versus NRT and/or differences in the study populations (i.e., younger, mostly female, Caucasian population in Lengsfeld et al. [129] versus older, predominantly African American, prediabetic or overweight male population in Yammine et al. [128]). Overall, these findings support further clinical trials of GLP-1R agonists in treatment-seeking smokers. Interestingly, preclinical studies showed that co-administration of liraglutide and nicotine had greater efficacy to decrease fat mass and body weight compared to either treatment alone (albeit in diet-induced obese mice that are not experiencing nicotine withdrawal), supporting a GLP-1R/NRT combinatorial approach to achieve the greatest effects on body weight [95]. Future clinical trials should identify efficacy and adverse effect profiles for potential GLP-1R agonist co-treatments to maximize patient compliance and reduction of body weight gain during nicotine withdrawal.

5.2.3. GLP-1R agonists and nicotine withdrawal-induced hyperphagia and body weight gain: neural mechanisms

Nicotine withdrawal-induced body weight gain involves both behavioral and metabolic changes [142]. Increased energy intake is a major factor contributing to nicotine withdrawal-induced body weight gain. Subjects who quit smoking increased their mean daily calorie consumption by 227 kcal, which explained 69 % of body weight gain observed at three months [17]. While clinical studies cannot identify the specific changes in feeding architecture that lead to increased caloric intake, pre-clinical studies showed that the effects of nicotine withdrawal on meal patterns were complex and included dynamic changes in meal size, meal number, and inter-meal interval [147,155157]. Overall, withdrawal-induced hyperphagia could be, in part, due to a reversal of the appetite suppressant effects of nicotine [15]. However, nicotine withdrawal leads to increased motivation for/consumption of highly palatable foods in humans and rodents, suggesting that alterations in homeostatic feeding do not fully explain withdrawal-induced hyperphagia [15,20,21,145,158,159]. Furthermore, the effects of nicotine cessation on food intake are variable between studies. Hyperphagia did not occur in several studies measuring intake of normal or bland chow in male and female rats during nicotine abstinence [38,147150]. However, hyperphagia was observed in studies that utilized a high fat diet or food restriction [38,146,160]. While route/dose of nicotine exposure and study design are undoubtedly a factor in the effects of nicotine withdrawal on hyperphagia, these findings suggest that nicotine withdrawal has a complicated reward value-enhancing effect on food that may be unmasked in the presence of high palatability or food scarcity [161,162].

The rewarding effects of nicotine and food are mediated by the mesolimbic dopamine reward system [163], and nicotine withdrawal alters dopamine signaling in the striatum [164,165]. GLP-1R agonists suppressed phasic dopamine responses to food-related cues [116], and pharmacological inhibition of dopamine receptors in the NAc prevented cue-induced reinstatement to sucrose seeking in drug-naïve rats [166]. Therefore, it is possible that GLP-1R agonists reduce central dopamine signaling in response to food-related cues during nicotine withdrawal, which may, in part, underlie its efficacy to reduce withdrawal-induced hyperphagia. Measuring the effects of GLP-1R agonists on VTA dopamine neuron responses to food cues during nicotine withdrawal could verify this potential mechanism. In addition to central dopamine signaling, preclinical studies showed that corticotropin-releasing factor (CRF) and hypothalamic CART regulated the effects of nicotine withdrawal on body weight [167,168]. Since GLP-1R agonists act directly on POMC/CART neurons in the arcuate nucleus of the hypothalamus to reduce food intake [169] and GLP-1-expressing neurons directly target paraventricular nucleus CRF neurons [170], these circuits should also be considered as potential targets by which GLP-1R agonists reduce nicotine withdrawal-induced hyperphagia and body weight gain. Nicotine withdrawal-induced body weight gain is not explained completely by increased caloric intake [171]. While a reduction in food intake is responsible for the majority of weight loss during GLP-1R agonist treatment, GLP-1R agonists also increase metabolism and energy expenditure through central pathways [79]. Therefore, it is likely that GLP-1R agonists also increase metabolism to prevent nicotine withdrawal-induced body weight gain. Given the complexity of the neural circuits underlying the effects of nicotine and nicotine withdrawal on food intake, GLP-1R agonists likely act on multiple systems to attenuate nicotine withdrawal-induced hyperphagia and body weight gain [15,142]. Further studies are also necessary to clarify whether GLP-1R agonists prevent or reverse the neural mechanisms underlying the effects of nicotine withdrawal on food intake and body weight gain or engage separate mechanisms that oppose the effects of nicotine withdrawal on these phenotypes. While the neural mechanisms underlying the efficacy of GLP-1R agonists remain unclear, the preclinical and clinical evidence discussed here support GLP-1R-based approaches to treating nicotine withdrawal-induced hyperphagia and weight gain.

5.3. GLP-1R agonists and nicotine withdrawal-induced cognitive deficits

Nicotine withdrawal is associated with decreased verbal and working memory in abstinent smokers [172,173], and cognitive impairments during abstinence predicted more rapid relapse to nicotine use [174]. Higher-order cognitive function is associated with the ability to exert “self-control” (i.e., motivate behaviors based on expected rewards and penalties) [175,176]. Therefore, in addition to withdrawal-induced cognitive deficits promoting relapse via negative reinforcement [135], dysregulation of higher-order cognitive systems may directly increase relapse vulnerability by weakening circuits that are necessary for maintaining abstinence [177]. Thus, enhancing cognitive function has emerged as a novel approach to treating NUD [177179]. Acetylcho-linesterase inhibitors – drugs like galantamine and donepezil that improve cognitive deficits by increasing cholinergic signaling in the brain [180] – improved cognitive performance during nicotine withdrawal in mice [181] and prevented the reinstatement of nicotine-seeking behavior in rats [182,183]. Consistent with these results, galantamine decreased smoking rates in treatment-seeking smokers [184]. FDA-approved NUD medications like varenicline and bupropion also improve cognitive function during nicotine abstinence [174,185,186] and decrease relapse to smoking [187,188]. These studies indicate that pharmacotherapies aimed at enhancing cognitive performance during withdrawal may promote long-term smoking abstinence.

Emerging evidence suggests that GLP-1R agonists improve cognitive function. In preclinical studies, GLP-1R agonists increased learning and memory in drug-naïve animals as well as in animal models of T2DM, neurodegenerative disease, and neuropathic pain [42,189196]. While clinical studies investigating the impact of GLP-1R agonists on cognitive function showed mixed results, several studies found that GLP-1R agonists improved cognitive performance in participants with prediabetes or T2DM [197201]. These studies support the efficacy of GLP-1R agonists to maintain and improve cognitive function across several disease states. However, no studies have investigated whether GLP-1R agonist treatment improves cognitive function during withdrawal following nicotine self-administration. Nicotine withdrawal increases markers of inflammation, and treatment with the nonsteroidal anti-inflammatory drug indomethican prevented nicotine withdrawal-induced deficits in novel object recognition in mice [202]. GLP-1R agonists also have neuroprotective/anti-inflammatory effects while improving cognitive performance in models of T2DM and dementia [190,191]. Together, these studies suggest that GLP-1R agonists may improve cognitive deficits during nicotine withdrawal, in part, by decreasing neuroinflammation. Importantly, individuals with T2DM are at higher risk of developing cognitive impairments [203]. Given that cognitive impairments are associated with higher risk of smoking relapse [177], GLP-1R agonists may be particularly effective at preventing withdrawal-induced cognitive impairments and decreasing smoking relapse in this patient population. Future studies should investigate whether GLP-1R agonists decrease neuroinflammation and improve cognitive function during nicotine withdrawal in the presence and absence of metabolic disease.

5.4. GLP-1R agonists and nicotine withdrawal-induced affective symptoms

Withdrawal from nicotine produces negative affective symptoms, including anxiety and depression [204]. Anxiety-like behaviors are assessed in rodents with a battery of behavioral tests including conditioned place aversion, open field test (OFT), elevated plus maze (EPM), and light-dark box, while depressive-like behaviors are assessed with the forced swim test (FST), and tail suspension test (TST), among others [131,205211]. Nicotine withdrawal increases anxiety- and depressive-like behaviors in these assays, supporting their validity to screen potential treatments for nicotine withdrawal phenotypes [205, 207,209,212].

While no studies have directly investigated the efficacy of GLP-1R agonists to prevent nicotine withdrawal-induced affective symptoms, GLP-1R agonists decreased anxiety- and depressive-like behaviors in drug-naïve animals. For example, repeated liraglutide administration decreased immobility in the FST, a measure of behavioral despair, in mice with and without chronic unpredictable stress experience [195]. In another study utilizing a model of corticosterone-induced depressive--like behaviors, repeated liraglutide decreased immobility in the TST and FST and increased time spent in the center of an open field [40]. However, liraglutide did not affect anxiety-like behavior in the EPM. These results suggest that GLP-1R agonists may have antidepressant-like effects, but their anxiolytic-like effects are unclear. It is important to note that while repeated liraglutide had no effects in corticosterone-naïve mice, the DPP-IV inhibitor sitagliptin decreased immobility time in both the FST and TST [196]. Therefore, liraglutide may only affect immobility time in animals already experiencing stress or exhibiting depressive-like behaviors. These studies only investigated the effects of liraglutide on depressive- and anxiety-like behaviors in male rodents. A recent study extended these findings to female rats and found that repeated liraglutide prevented ovariectomy-induced despair and anxiety-like behavior as measured by FST, OFT, and EPM [213]. Together, these findings support the efficacy of liraglutide to decrease depressive- and anxiety-like behaviors in both male and female rodents. Repeated dulaglutide treatment improved chronic social defeat stress-induced anxiety- and depressive-like behaviors and improved biomarkers of inflammation, identifying a potential mechanism underlying the effects of GLP-1R agonists on anxiety- and depressive-like behaviors [214]. Interestingly, repeated Exendin-4 attenuated lipopolysaccharide-induced depressive-like behavior but did not affect markers of inflammation [215]. These results suggest that GLP-1R agonists may act via separate mechanisms to alleviate depressive-like symptoms depending on the agonist and/or paradigm studied, which is supported by previous studies demonstrating differences in the neuroprotective effects of GLP-1R agonists [216]. In sum, preclinical studies demonstrated antidepressant-like and anxiolytic-like effects of GLP-1R agonists in drug-naïve animals. Therefore, future studies should investigate whether GLP-1R agonists decrease affective symptoms like anxiety and depression during nicotine withdrawal.

GLP-1R agonists act on neural circuits implicated in the pathophysiology of depression. Depression is associated with neuroinflammation, deficits in neuroplasticity, and dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis [217219]. In preclinical studies of anxiety- and depressive-like behaviors, inflammatory biomarkers, hippocampal LTP, adult neurogenesis, and levels of the stress hormone adrenocorticotropic hormone were all normalized by GLP-1R agonist treatment [40,41,214]. Therefore, these central mechanisms may mediate the effects of GLP-1R agonists on nicotine withdrawal symptoms. Future studies should investigate the effects of GLP-1R agonists on neuroplasticity, neuroinflammation, and stress systems during NUD and their relationship to depression- and anxiety-like behaviors.

HPA axis dysregulation is not only associated with the development of depression but also with the affective symptoms of nicotine withdrawal. Smokers have elevated cortisol levels compared to non-smokers [220,221] and nicotine withdrawal leads to a rebound decrease in cortisol/corticosterone levels in both human smokers and animal models [222,223]. Somewhat counterintuitively, lower cortisol levels during nicotine withdrawal were associated with increased craving, self-reported withdrawal symptoms, and subjective stress [222,224]. Furthermore, nicotine withdrawal decreased cortisol/corticosterone levels in response to acute stressors [223,225,226]. These findings indicate that reduced HPA axis function during withdrawal may be related to smoking cessation-induced stress and depression [222,223]. Therefore, treatments that normalize HPA axis function during nicotine withdrawal may help to prevent depressive-like behaviors. Importantly, GLP-1R agonists increased HPA axis function/corticosterone release in healthy and streptozotocin-induced-diabetic rats and increased cortisol release in drug-naïve humans (both T2DM participants and healthy controls), suggesting that GLP-1R agonists could help to ameliorate HPA axis dysregulation during nicotine withdrawal [227,228]. Future studies should investigate whether GLP-1R agonists reverse the effects of nicotine withdrawal on HPA axis function and determine whether changes in HPA axis activity during withdrawal are correlated with self-report scale measurements of nicotine craving, withdrawal, and stress during GLP-1R agonist treatment.

Depression is also associated with dysregulation of the gut-brain axis and energy homeostasis [41,229231]. Smoking cessation alters gut microbiota composition and diversity [232], and nicotine disrupts mitochondrial function [233]. Therefore, disruption of the gut-brain axis and energy homeostasis may underlie, in part, the affective symptoms of nicotine withdrawal. GLP-1R agonists modulated gut microbiota composition and diversity and improved mitochondrial biogenesis and function [41,234236]. Therefore, GLP-1R agonists may alter the microbiome and mitochondrial function to prevent depression-like behaviors during nicotine withdrawal. Future studies should measure the effects of GLP-1R agonists on mitochondrial function and the microbiome during nicotine withdrawal to identify whether GLP-1R agonists improve withdrawal-induced deficits in these systems. Given the high comorbidity of both depression and smoking with metabolic diseases like T2DM, it is critical to understand how altered function of the gut-brain axis and dysregulated energy homeostasis contribute to depressive-like symptoms during nicotine withdrawal [15,237,238]. Although further research is needed, the current literature suggests that GLP-1R agonists could be targeted towards this patient population to improve nicotine withdrawal-induced depression.

While there are no published clinical trials investigating the effects of GLP-1R agonists on depression or anxiety, there are several retrospective observational studies that measured these outcomes. One population-based, longitudinal study identified a decreased risk of depression and anxiety in participants with T2DM treated with GLP-1R agonists [239]. Significant effects were observed in female, but not male participants. These effects were also specific to dulaglutide, with no effects observed in subjects treated with liraglutide or exenatide. Risk of anxiety was decreased more than risk of depression. Another recent clinical study found that low doses of GLP-1R agonists and DPP-IV inhibitors were associated with a lower risk of depression in T2DM participants [240]. Furthermore, a meta-analysis demonstrated that GLP-1R agonists reduced depression severity scores, with a stronger effect in diabetic participants [241]. However, one analysis of participants with T2DM found no effects of GLP-1R agonists or DPP-IV inhibitors on risk of a new diagnosis of depression or episodes of self-harm [242]. Furthermore, a retrospective population-based study found no effects of GLP-1R agonists on depression [243]. Importantly, a recent retrospective study found that GLP-1R agonists decreased suicidal ideation compared to non-GLP-1R agonist anti-obesity or anti-diabetes medications across large cohorts of patients with overweight/obesity or T2DM [244]. These effects were consistent across sex, age, and ethnicity stratification. Overall, while there are promising results demonstrating potential antidepressant and anxiolytic effects of GLP-1R agonists, the lack of controlled studies and mixed results make it impossible to draw firm conclusions about the efficacy of these drugs to decrease mood disorders [245]. Furthermore, no studies have investigated whether GLP-1R agonists decrease depression and anxiety in participants with NUD. Future randomized, blinded, placebo-controlled trials investigating the therapeutic benefit of GLP-1R agonists in participants with NUD should include affective symptoms of withdrawal as primary outcomes to better understand their overall effects on anxiety and depression in abstinent smokers.

5.5. GLP-1R agonists and cardiovascular disease

Smoking is a major risk factor for atherosclerotic cardiovascular disease [246]. While quitting smoking decreases the incidence of cardiovascular disease [247], decreasing tobacco consumption can actually increase cardiovascular disease risk factors like blood pressure, high-density lipoprotein cholesterol, body weight, and waist-to-hip ratio [248]. Furthermore, heavy smokers (≥ 20 pack-years) continue to have an elevated risk of cardiovascular disease 10–15 years after smoking cessation [249]. Unfortunately, nicotine withdrawal-induced body weight gain can prevent the beneficial effects of quitting on markers of cardiovascular health [250] and elevate the short-term risk of T2DM [251]. This is particularly concerning given the elevated risk of cardiovascular disease in individuals with T2DM [252]. GLP-1R agonists including liraglutide and semaglutide had positive effects on cardiovascular outcomes in participants with T2DM [60]. Specifically, participants with T2DM treated with liraglutide had a 13 % reduction in major adverse cardiovascular events compared to placebo [253]. This reduction was likely due to decreased atherosclerosis, as changes in HbA1c, body weight, and blood pressure were not significant enough to mediate the effect [60,253]. Another clinical study found a 26 % reduction in major adverse cardiovascular events in participants with T2DM treated with semaglutide compared to placebo [254]. Semaglutide also improved cardiometabolic risk factors including waist circumference, systolic/diastolic blood pressure, fasting plasma glucose, and lipids in individuals with overweight/obesity without T2DM [255]. While the mechanism(s) underlying these effects is unclear, GLP-1R agonists may reduce atherosclerosis via decreasing inflammation [256]. Together, these findings demonstrate improved cardiovascular outcomes after GLP-1R agonist treatment and suggest that GLP-1R agonists may exert cardioprotective effects during nicotine withdrawal. However, it is important to note that these studies were all conducted with participants that were overweight/obese or had T2DM, so the cardioprotective efficacy of GLP-1R agonists may differ in participants without metabolic diseases. Nonetheless, these results demonstrate reduced cardiovascular risk in a highly clinically relevant population. Future studies should investigate the effects of GLP-1R agonists on cardiovascular risk for participants with and without metabolic disease. These results could identify a novel benefit of GLP-1R agonist treatment that may be critical for individuals at risk of negative cardiovascular outcomes during nicotine withdrawal.

6. Ongoing clinical trials and areas for future study

Overall, pilot clinical studies validate preclinical reports and suggest that GLP-1R agonists attenuate nicotine withdrawal-induced body weight gain [128,129]. While the evidence is not as strong, these studies also suggest that GLP-1R agonists may reduce nicotine craving and smoking relapse. However, studies published to date are limited in number and have relatively small sample sizes that limit the conclusions that can be drawn from their results. More robust findings from larger studies are necessary to fully understand the clinical potential of GLP-1R agonists to treat NUD. There are recently completed and ongoing clinical trials that will improve our current understanding of GLP-1R agonists as potential NUD pharmacotherapies (Table 2). One recently completed double-blind, parallel arm study measured the effects of 32 weeks of liraglutide treatment (escalating doses of 0.6–3.0 mg, s.c.) on smoking abstinence, calorie consumption, and body weight change in overweight and obese smokers (ClinicalTrials.gov ID NCT03712098). Participants also underwent eight sessions of smoking cessation behavioral counseling. By measuring calorie consumption during liraglutide treatment and nicotine withdrawal, this study will determine if reduced caloric intake is the primary mechanism by which GLP-1R agonists reduce body weight gain during withdrawal. Another study is currently recruiting participants to investigate the effects of nine weeks of escalating weekly doses of semaglutide (0.25–1.0 mg, s.c.) on smoking behaviors (ClinicalTrials.gov ID NCT05530577). Specifically, this study will investigate changes in resistance to smoking relapse after overnight abstinence and tobacco smoking after eight weeks of treatment. Changes in cigarette craving during exposure to smoking-related conditioned cues and subjective responses to cigarette smoking will also be measured. Finally, this study will assess changes in daily cigarettes smoked, body weight, and HbA1c levels from baseline to the study endpoint at week 10. No clinical trial has measured the efficacy of GLP-1R agonist monotherapy alone to treat NUD. Therefore, this study will provide our first insights into the efficacy of GLP-1R agonist monotherapy for NUD. Yammine et al. are also following up on their pilot trial in a randomized, double-blind, placebo-controlled study that is currently recruiting study participants (2021). This study will determine the effects of 14 weeks of exenatide (2 mg) on nicotine withdrawal-induced body weight gain in overweight and/or prediabetic participants (ClinicalTrials.gov ID NCT05610800). Participants will also receive smoking cessation counseling and de-escalating doses of NRT during the active treatment phase. Abstinence rates and body weight change will be assessed at 14 weeks and 26 weeks after treatment initiation. Neural responses to visual stimuli will also be measured to identify the effects of GLP-1R agonism on neural correlates of craving [257]. Overall, this study will provide more information about the neural mechanisms underlying the effects of GLP-1R agonists on smoking relapse. It will also determine the efficacy of GLP-1R agonists to reduce withdrawal-induced body weight gain and increase abstinence in a larger sample population. More recently, Yammine et al. registered a study (not yet recruiting) investigating the effects of semaglutide (0.24–2.4 mg, s.c.) on post-smoking cessation weight management (ClinicalTrials.gov ID NCT06173778). In addition to semaglutide, participants will also receive smoking cessation counseling and de-escalating doses of NRT. This study will expand our understanding of the efficacy of newer generations of GLP-1R agonists for preventing nicotine withdrawal-induced weight gain.

Given that overweight and obese smokers are at greater risk for body weight gain during nicotine withdrawal [19], it is encouraging that exenatide decreased withdrawal-induced body weight gain in smokers with prediabetes and/or overweight [128,258]. However, future studies should also investigate the effects of GLP-1R agonists on hyperphagia and body weight gain in other subgroups including smokers with T2DM. This population is particularly important to study given their higher risk for worsening glycemic control during smoking cessation and that traditional pharmacotherapies for smoking cessation may be less effective for them [258261]. Preclinical studies supporting GLP-1R agonist treatment of nicotine withdrawal-induced hyperphagia and body weight gain utilized a high fat diet [38,146]. The current clinical literature does not measure or manipulate food type in their analyses of GLP-1R efficacy. Therefore, future studies should also investigate whether GLP-1R agonists retain their efficacy to reduce nicotine withdrawal-induced hyperphagia and body weight gain when participants consume a healthy diet versus a western diet high in fats and sugars. It is also important to continue to investigate the effects of newer generation GLP-1R agonists like semaglutide on NUD and associated withdrawal symptoms, given that GLP-1R agonists have differences in pharmacodynamics, pharmacokinetics, and brain penetrance [63,75,82]. Future studies should also compare the efficacy of GLP-1R agonist monotherapy alone to combinatorial treatments with FDA-approved pharmacotherapies including NRT and varenicline. While one study found a general effect of GLP-1R agonist treatment on nicotine withdrawal symptoms as measured by the Wisconsin Scale of Withdrawal Symptoms, a 28 item questionnaire evaluating anger, anxiety, concentration, craving, hunger, sadness, and sleep, future studies should measure the effects of GLP-1R agonists on specific symptoms of withdrawal (e.g., separate measurements of somatic, cognitive, and affective symptoms) [128]. These measurements would allow precision treatment with GLP-1R agonists alone or in combination with other therapies to target the most relevant symptoms for each patient. Interestingly, there are several anecdotal reports of individuals who were taking semaglutide for weight loss that unexpectedly quit smoking due to cigarettes suddenly tasting “repulsive” or “disgusting” [262,263]. Given that GLP-1 is synthesized in taste buds, GLP-1Rs are expressed on taste bud afferent nerve fibers, and GLP-1 signaling affects taste perception [264266], it is possible that, in addition to their central effects, GLP-1R agonists may act directly at taste buds to alter smoking behavior. Indeed, constitutive GLP-1R knockout decreased taste responses to sweeteners and may increase sour taste sensitivity, effects that could alter the flavor profile of tobacco smoke and reduce smoking behaviors [266]. Future studies should investigate changes in taste sensitivity during GLP-1R agonist treatment and the relationship between taste sensitivity and nicotine abstinence to further understand this potential mechanism.

7. Concluding remarks

A small but growing preclinical literature indicates that GLP-1R agonists attenuate voluntary nicotine taking- and seeking-behaviors as well as nicotine withdrawal-induced hyperphagia and body weight gain in rodents. Consistent with these findings, clinical studies indicate that GLP-1R agonists reduce nicotine withdrawal-induced hyperphagia and body weight gain, two major obstacles to quitting smoking and achieving long-term abstinence in male and female smokers. Despite these promising results, the effects of GLP-1R agonists on nicotine craving and abstinence in human smokers have been mixed. While emerging studies in drug-naïve animals suggests that GLP-1R agonists may improve cognitive deficits, depression, and/or anxiety, there are no studies directly measuring these effects in nicotine-exposed laboratory animals or human smokers. The neural mechanisms underlying the effects of GLP-1R agonists on nicotine-mediated behaviors are not clear, and likely involve neural circuits mediating reward and aversion. To determine the exact mechanisms underlying the efficacy of GLP-1R agonists, future studies should investigate the effects of GLP-1R agonists on nicotine-induced changes in neurotransmitter signaling and synaptic plasticity in nuclei regulating nicotine taking and seeking including the VTA, NAc, MHb, IPN, hypothalamus, and hippocampus. Future studies should also investigate the effects of GLP-1R agonist treatment on neuroinflammation and HPA axis function and their relationship to cognitive and affective symptoms during nicotine withdrawal. These experiments would clarify potential mechanisms underlying the efficacy of GLP-1R agonists to improve cognitive deficits and produce anxiolytic- and/or antidepressant-like responses during nicotine withdrawal. Finally, clinical studies establishing which GLP-1R agonists and treatment protocols (e.g., monotherapy versus combinatorial treatments) are most effective in promoting long-term smoking abstinence while minimizing adverse effects will enhance their potential as NUD pharmacotherapies. Understanding the specific patient populations and nicotine withdrawal symptoms that GLP-1R agonists most effectively treat may also facilitate their use as customized, efficacious treatments for NUD.

Acknowledgments

This work was supported by the following grants from the National Institutes of Health: R01 DA037897 and R21 DA045792 (H.D.S.) and training grant T32 DA028874 and fellowship F31 DA058451 (R.J.H.). The authors would also like to thank Dr. Matthew Hayes for his critical reading of an earlier draft of this manuscript.

Footnotes

CRediT authorship contribution statement

Rae J. Herman: Writing – review & editing, Writing – original draft. Heath D. Schmidt: Writing – review & editing, Writing – original draft.

Declaration of competing interest

The authors declare no competing financial interests.

Data availability

No data was used for the research described in the article.

References

  • [1].CDC, Smoking Cessation: A Report of the Surgeon General, CDC Center for Disease Control and Prevention, 2020. [Google Scholar]
  • [2].Cornelius ME, Loretan CG, Jamal A, Davis Lynn BC, Mayer M, Alcantara IC, et al. , Tobacco product use among adults - United States, 2021, MMWR Morb. Mortal. Wkly. Rep 72 (2023) 475–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Cooper M, Park-Lee E, Ren C, Cornelius M, Jamal A, Cullen KA, Notes from the field: e-cigarette use among middle and high school students - United States, 2022, MMWR. Morb. Mortal. Wkly. Rep 71 (2022) 1283–1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Anic GM, Sawdey MD, Jamal A, Trivers KF, Frequency of use among middle and high school student tobacco product users - United States, 2015–2017, MMWR Morb. Mortal. Wkly. Rep 67 (2018) 1353–1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Soneji S, Errors in data input in meta-analysis on association between initial use of e-cigarettes and subsequent cigarette smoking among adolescents and young adults, JAMA Pediatr 172 (2018) 92–93. [DOI] [PubMed] [Google Scholar]
  • [6].Soneji S, Barrington-Trimis JL, Wills TA, Leventhal AM, Unger JB, Gibson LA, et al. , Association between initial use of e-cigarettes and subsequent cigarette smoking among adolescents and young adults: a systematic review and meta-analysis, JAMA Pediatr 171 (2017) 788–797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Taylor DH Jr., Hasselblad V, Henley SJ, Thun MJ, Sloan FA, Benefits of smoking cessation for longevity, Am. J. Public Health 92 (2002) 990–996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Babb S, Malarcher A, Schauer G, Asman K, Jamal A, Quitting smoking among adults - United States, 2000–2015, MMWR Morb. Mortal. Wkly. Rep 65 (2017) 1457–1464. [DOI] [PubMed] [Google Scholar]
  • [9].Gentzke AS, Cornelius TW,M, et al. , Tobacco product use and associated factors among middle and high school students — National Youth Tobacco Survey, United States, 2021, MMWR Surveill. Summ 77 (2022) 1–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Stolerman IP, Jarvis MJ, The scientific case that nicotine is addictive, Psychopharmacology (Berl.) 117 (1995) 2–10. [DOI] [PubMed] [Google Scholar]
  • [11].Dai H, Prevalence and factors associated with youth vaping cessation intention and quit attempts, Pediatrics (2021) 148. [DOI] [PubMed] [Google Scholar]
  • [12].Soule EK, Lee JGL, Egan KL, Bode KM, Desrosiers AC, Guy MC, et al. , I cannot live without my vape”: electronic cigarette user-identified indicators of vaping dependence, Drug Alcohol Depend 209 (2020) 107886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Klesges RC, Brown K, Pascale RW, Murphy M, Williams E, Cigrang JA, Factors associated with participation, attrition, and outcome in a smoking cessation program at the workplace, Health Psychol 7 (1988) 575–589. [DOI] [PubMed] [Google Scholar]
  • [14].Meyers AW, Klesges RC, Winders SE, Ward KD, Peterson BA, Eck LH, Are weight concerns predictive of smoking cessation? A prospective analysis, J. Consult. Clin. Psychol 65 (1997) 448–452. [DOI] [PubMed] [Google Scholar]
  • [15].Audrain-McGovern J, Benowitz NL, Cigarette smoking, nicotine, and body weight, Clin. Pharmacol. Ther 90 (2011) 164–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Chao AM, Wadden TA, Ashare RL, Loughead J, Schmidt HD, Tobacco smoking, eating behaviors, and body weight: a review, Curr. Addict. Rep 6 (2019) 191–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Stamford BA, Matter S, Fell RD, Papanek P, Effects of smoking cessation on weight gain, metabolic rate, caloric consumption, and blood lipids, Am. J. Clin. Nutr 43 (1986) 486–494. [DOI] [PubMed] [Google Scholar]
  • [18].Williamson DF, Madans J, Anda RF, Kleinman JC, Giovino GA, Byers T, Smoking cessation and severity of weight gain in a national cohort, N. Engl. J. Med 324 (1991) 739–745. [DOI] [PubMed] [Google Scholar]
  • [19].Bush T, Lovejoy JC, Deprey M, Carpenter KM, The effect of tobacco cessation on weight gain, obesity and diabetes risk, Obesity (Silver Spring) 24 (2016) 1834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Spring B, Pagoto S, McChargue D, Hedeker D, Werth J, Altered reward value of carbohydrate snacks for female smokers withdrawn from nicotine, Pharmacol. Biochem. Behav 76 (2003) 351–360. [DOI] [PubMed] [Google Scholar]
  • [21].Perkins KA, Epstein LH, Sexton JE, Pastor S, Effects of smoking cessation on consumption of alcohol and sweet, high-fat foods, J. Subst. Abuse 2 (1990) 287–297. [DOI] [PubMed] [Google Scholar]
  • [22].Janzon E, Hedblad B, Berglund G, Engström G, Changes in blood pressure and body weight following smoking cessation in women, J. Intern. Med 255 (2004) 266–272. [DOI] [PubMed] [Google Scholar]
  • [23].Chinn S, Jarvis D, Melotti R, Luczynska C, Ackermann-Liebrich U, Antó JM, et al. , Smoking cessation, lung function, and weight gain: a follow-up study, Lancet 365 (2005) 1629–1635, discussion 00–1. [DOI] [PubMed] [Google Scholar]
  • [24].Yeh HC, Duncan BB, Schmidt MI, Wang NY, Brancati FL, Smoking, smoking cessation, and risk for type 2 diabetes mellitus: a cohort study, Ann. Intern. Med 152 (2010) 10–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Benowitz NL, Cigarette smoking and cardiovascular disease: pathophysiology and implications for treatment, Prog. Cardiovasc. Dis 46 (2003) 91–111. [DOI] [PubMed] [Google Scholar]
  • [26].Chiolero A, Faeh D, Paccaud F, Cornuz J, Consequences of smoking for body weight, body fat distribution, and insulin resistance, Am. J. Clin. Nutr 87 (2008) 801–809. [DOI] [PubMed] [Google Scholar]
  • [27].Mills EJ, Wu P, Lockhart I, Thorlund K, Puhan M, Ebbert JO, Comparisons of high-dose and combination nicotine replacement therapy, varenicline, and bupropion for smoking cessation: a systematic review and multiple treatment meta-analysis, Ann. Med 44 (2012) 588–597. [DOI] [PubMed] [Google Scholar]
  • [28].Farley AC, Hajek P, Lycett D, Aveyard P, Interventions for preventing weight gain after smoking cessation, Cochrane Database Syst. Rev 1 (2012) CD006219. [DOI] [PubMed] [Google Scholar]
  • [29].Parsons AC, Shraim M, Inglis J, Aveyard P, Hajek P, Interventions for preventing weight gain after smoking cessation, Cochrane Database Syst. Rev (2009). [DOI] [PubMed] [Google Scholar]
  • [30].Davies M, Færch L, Jeppesen OK, Pakseresht A, Pedersen SD, Perreault L, et al. , Semaglutide 2·4 mg once a week in adults with overweight or obesity, and type 2 diabetes (STEP 2): a randomised, double-blind, double-dummy, placebo-controlled, phase 3 trial, Lancet 397 (2021) 971–984. [DOI] [PubMed] [Google Scholar]
  • [31].Lewis AL, McEntee N, Holland J, Patel A, Development and approval of rybelsus (oral semaglutide): ushering in a new era in peptide delivery, Drug Deliv. Transl. Res 12 (2022) 1–6. [DOI] [PubMed] [Google Scholar]
  • [32].Wilding JPH, Batterham RL, Calanna S, Davies M, Van Gaal LF, Lingvay I, et al. , Once-weekly semaglutide in adults with overweight or obesity, N. Engl. J. Med 384 (2021) 989–1002. [DOI] [PubMed] [Google Scholar]
  • [33].Jerlhag E, Gut-brain Axis and Addictive Disorders: A Review With Focus On Alcohol and Drugs of Abuse, Pharmacology and Therapeutics: Elsevier Inc, 2019, pp. 1–14. [DOI] [PubMed] [Google Scholar]
  • [34].Merkel R, Moreno A, Zhang Y, Herman R, Ben Nathan J, Zeb S, et al. , A novel approach to treating opioid use disorders: dual agonists of glucagon-like peptide-1 receptors and neuropeptide Y(2) receptors, Neurosci. Biobehav. Rev 131 (2021) 1169–1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Hayes MR, Schmidt HD, GLP-influences Food and Drug Reward. Current Opinion in Behavioral Sciences, Elsevier Ltd, 2016, pp. 66–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Jerlhag E, The therapeutic potential of glucagon-like peptide-1 for persons with addictions based on findings from preclinical and clinical studies, Front. Pharmacol (2023) 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Hernandez NS, Schmidt HD, Central GLP-1 receptors: novel molecular targets for cocaine use disorder, Physiol. Behav 206 (2019) 93–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Herman RJ, Hayes MR, Audrain-McGovern J, Ashare RL, Schmidt HD, Liraglutide attenuates nicotine self-administration as well as nicotine seeking and hyperphagia during withdrawal in male and female rats, Psychopharmacology (Berl) 240 (6) (2023) 1373–1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Tuesta LM, Chen Z, Duncan A, Fowler CD, Ishikawa M, Lee BR, et al. , GLP-1 acts on habenular avoidance circuits to control nicotine intake, Nat. Neurosci 20 (2017) 708–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Weina H, Yuhu N, Christian H, Birong L, Feiyu S, Le W, Liraglutide attenuates the depressive- and anxiety-like behaviour in the corticosterone induced depression model via improving hippocampal neural plasticity, Brain Res 1694 (2018) 55–62. [DOI] [PubMed] [Google Scholar]
  • [41].Detka J, Głombik K, Insights into a possible role of glucagon-like peptide-1 receptor agonists in the treatment of depression, Pharmacol. Rep 73 (2021) 1020–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Trammell TS, Henderson NL, Madkour HS, Stanwood GD, Graham DL, GLP-1R activation alters performance in cognitive tasks in a sex-dependent manner, Neurol. Sci 42 (2021) 2911–2919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Larsen PJ, Tang-Christensen M, Holst JJ, Ørskov C, Distribution of glucagon-like peptide-1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem, Neuroscience 77 (1997) 257–270. [DOI] [PubMed] [Google Scholar]
  • [44].Holt MK, Richards JE, Cook DR, Brierley DI, Williams DL, Reimann F, et al. , Preproglucagon neurons in the nucleus of the solitary tract are the main source of brain GLP-1, mediate stress-induced hypophagia, and limit unusually large intakes of food, Diabetes 68 (2019) 21–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Baggio LL, Drucker DJ, Biology of incretins: GLP-1 and GIP, Gastroenterology 132 (2007) 2131–2157. [DOI] [PubMed] [Google Scholar]
  • [46].Hayes MR, Leichner TM, Zhao S, Lee GS, Chowansky A, Zimmer D, et al. , Intracellular signals mediating the food intake-suppressive effects of hindbrain glucagon-like peptide-1 receptor activation, Cell Metab 13 (2011) 320–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Holst JJ, The physiology of glucagon-like peptide 1, Physiol. Rev 87 (2007) 1409–1439. [DOI] [PubMed] [Google Scholar]
  • [48].Merchenthaler I, Lane M, Shughrue P, Distribution of pre-pro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system, J. Comp. Neurol 403 (1999) 261–280. [DOI] [PubMed] [Google Scholar]
  • [49].Fletcher MM, Halls ML, Christopoulos A, Sexton PM, Wootten D, The complexity of signalling mediated by the glucagon-like peptide-1 receptor, Biochem. Soc. Trans 44 (2016) 582–588. [DOI] [PubMed] [Google Scholar]
  • [50].Müller TD, Finan B, Bloom SR, D’Alessio D, Drucker DJ, Flatt PR, et al. , Glucagon-Like Peptide 1 (GLP-1), Molecular Metabolism: Elsevier GmbH, 2019, pp. 72–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Wootten D, Reynolds CA, Smith KJ, Mobarec JC, Koole C, Savage EE, et al. , The extracellular surface of the GLP-1 receptor is a molecular trigger for biased agonism, Cell 165 (2016) 1632–1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Koole C, Wootten D, Simms J, Savage EE, Miller LJ, Christopoulos A, et al. , Second extracellular loop of human glucagon-like peptide-1 receptor (GLP-1R) differentially regulates orthosteric but not allosteric agonist binding and function, J. Biol. Chem 287 (2012) 3659–3673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Koole C, Wootten D, Simms J, Valant C, Sridhar R, Woodman OL, et al. , Allosteric ligands of the glucagon-like peptide 1 receptor (GLP-1R) differentially modulate endogenous and exogenous peptide responses in a pathway-selective manner: implications for drug screening, Mol. Pharmacol 78 (2010) 456–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Jones B, The therapeutic potential of GLP-1 receptor biased agonism, Br. J. Pharmacol 179 (2022) 492–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Jones B, Buenaventura T, Kanda N, Chabosseau P, Owen BM, Scott R, et al. , Targeting GLP-1 receptor trafficking to improve agonist efficacy, Nat. Commun 9 (2018) 1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Kolterman OG, Buse JB, Fineman MS, Gaines E, Heintz S, Bicsak TA, et al. , Synthetic exendin-4 (exenatide) significantly reduces postprandial and fasting plasma glucose in subjects with type 2 diabetes, J. Clin. Endocrinol. Metab 88 (2003) 3082–3089. [DOI] [PubMed] [Google Scholar]
  • [57].Eng J, Kleinman WA, Singh L, Singh G, Raufman JP, Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom. Further evidence for an exendin receptor on dispersed acini from guinea pig pancreas, J. Biol. Chem 267 (1992) 7402–7405. [PubMed] [Google Scholar]
  • [58].Anderson J, The pharmacokinetic properties of glucagon-like peptide-1 receptor agonists and their mode and mechanism of action in patients with type 2 diabetes, J. Fam. Pract 67 (2018) S8–s13. [PubMed] [Google Scholar]
  • [59].Li Y, Vaughan KL, Tweedie D, Jung J, Kim HK, Choi HI, et al. , Pharmacokinetics of exenatide in nonhuman primates following its administration in the form of sustained-release PT320 and bydureon, Sci. Rep 9 (2019) 17208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Knudsen LB, Lau J, The discovery and development of liraglutide and semaglutide, Front. Endocrinol. (Lausanne) (2019) 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Crane J, McGowan B, The GLP-1 Agonist, Liraglutide, As a Pharmacotherapy For Obesity, 7, Ther Adv Chronic Dis, 2016, pp. 92–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Nuffer W, Chapter 5 - pharmacologic agents chapter for abdominal obesity, in: Watson RR (Ed.), Nutrition in the Prevention and Treatment of Abdominal Obesity (Second Edition), Academic Press, 2019, pp. 51–66. [Google Scholar]
  • [63].Meier JJ, GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus, Nat. Rev. Endocrinol 8 (2012) 728–742. [DOI] [PubMed] [Google Scholar]
  • [64].Sposito AC, Berwanger O, de Carvalho LSF, K Saraiva JF, GLP-1RAs in type 2 diabetes: mechanisms that underlie cardiovascular effects and overview of cardiovascular outcome data, Cardiovasc. Diabetol 17 (2018) 157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Sharma D, Verma S, Vaidya S, Kalia K, Tiwari V, Recent updates on GLP-1 agonists: current advancements & challenges, Biomed. Pharmacother 108 (2018) 952–962. [DOI] [PubMed] [Google Scholar]
  • [66].Kapitza C, Nosek L, Jensen L, Hartvig H, Jensen CB, Flint A, Semaglutide, a once-weekly human GLP-1 analog, does not reduce the bioavailability of the combined oral contraceptive, ethinylestradiol/levonorgestrel, J. Clin. Pharmacol 55 (2015) 497–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Marbury TC, Flint A, Jacobsen JB, Derving Karsbøl J, Lasseter K, Pharmacokinetics and tolerability of a single dose of semaglutide, a human glucagon-like peptide-1 analog, in subjects with and without renal impairment, Clin. Pharmacokinet 56 (2017) 1381–1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Nauck MA, Petrie JR, Sesti G, Mannucci E, Courrèges J-P, Lindegaard ML, et al. , A phase 2, randomized, dose-finding study of the novel once-weekly human GLP-1 analog, semaglutide, compared with placebo and open-label liraglutide in patients with type 2 diabetes, Diabetes Care 39 (2015) 231–241. [DOI] [PubMed] [Google Scholar]
  • [69].Ahmann AJ, Capehorn M, Charpentier G, Dotta F, Henkel E, Lingvay I, et al. , Efficacy and safety of once-weekly semaglutide versus exenatide ER in subjects with type 2 diabetes (SUSTAIN 3): a 56-week, open-label, randomized clinical trial, Diabetes Care 41 (2017) 258–266. [DOI] [PubMed] [Google Scholar]
  • [70].Lingvay I, Desouza CV, Lalic KS, Rose L, Hansen T, Zacho J, et al. , A 26-week randomized controlled trial of semaglutide once daily versus liraglutide and placebo in patients with type 2 diabetes suboptimally controlled on diet and exercise with or without metformin, Diabetes Care 41 (2018) 1926–1937. [DOI] [PubMed] [Google Scholar]
  • [71].U. Food, D. Administration, FDA Approves Novel, Dual-Targeted Treatment For Type 2 Diabetes, FDA News Release, 2022. [Google Scholar]
  • [72].Frías JP, Davies MJ, Rosenstock J, Pérez Manghi FC, Fernandez Landó L, Bergman BK, et al. , Tirzepatide versus semaglutide once weekly in patients with type 2 diabetes, N. Engl. J. Med 385 (2021) 503–515. [DOI] [PubMed] [Google Scholar]
  • [73].Finan B, Ma T, Ottaway N, Müller TD, Habegger KM, Heppner KM, et al. , Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans, Sci. Transl. Med 5 (2013), 209ra151. [DOI] [PubMed] [Google Scholar]
  • [74].Nauck MA, Quast DR, Wefers J, Meier JJ, GLP-1 receptor agonists in the treatment of type 2 diabetes – state-of-the-art, Mol. Metab 46 (2021) 101102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Uccellatore A, Genovese S, Dicembrini I, Mannucci E, Ceriello A, Comparison review of short-acting and long-acting glucagon-like peptide-1 receptor agonists, Diabetes Ther 6 (2015) 239–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Kyriacou A, Ahmed AB, Exenatide use in the management of type 2 diabetes mellitus, Pharmaceuticals. (Basel) 3 (2010) 2554–2567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Xue X, Ren Z, Zhang A, Yang Q, Zhang W, Liu F, Efficacy and safety of once-weekly glucagon-like peptide-1 receptor agonists compared with exenatide and liraglutide in type 2 diabetes: a systemic review of randomised controlled trials, Int. J. Clin. Pract 70 (2016) 649–656. [DOI] [PubMed] [Google Scholar]
  • [78].Trujillo JM, Nuffer W, Smith BA, GLP-1 receptor agonists: an updated review of head-to-head clinical studies, Ther. Adv. Endocrinol. Metab 12 (2021) 2042018821997320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Drucker DJ, Mechanisms of action and therapeutic application of glucagon-like peptide-1, Cell Metab 27 (2018) 740–756. [DOI] [PubMed] [Google Scholar]
  • [80].Sisley S, Gutierrez-Aguilar R, Scott M, D’Alessio DA, Sandoval DA, Seeley RJ, Neuronal GLP1R mediates liraglutide’s anorectic but not glucose-lowering effect, J. Clin. Invest 124 (2014) 2456–2463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Kanoski SE, Fortin SM, Arnold M, Grill HJ, Hayes MR, Peripheral and central GLP-1 receptor populations mediate the anorectic effects of peripherally administered GLP-1 receptor agonists, liraglutide and exendin-4, Endocrinology 152 (2011) 3103–3112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Gabery S, Salinas CG, Paulsen SJ, Ahnfelt-Rønne J, Alanentalo T, Baquero AF, et al. , Semaglutide lowers body weight in rodents via distributed neural pathways, JCI Insight (2021) 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Piguet O, Peterśen A, Yin Ka Lam B, Gabery S, Murphy K, Hodges JR, et al. , Eating and hypothalamus changes in behavioral-variant frontotemporal dementia, Ann. Neurol 69 (2011) 312–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Williams DL, Baskin DG, Schwartz MW, Evidence that intestinal glucagon-like peptide-1 plays a physiological role in satiety, Endocrinology 150 (2009) 1680–1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Buller S, Blouet C, Brain access of incretins and incretin receptor agonists to their central targets relevant for appetite suppression and weight loss, Am. J. Physiol.-Endocrinol. Metabol 326 (2024). E472–E80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Adams JM, Pei H, Sandoval DA, Seeley RJ, Chang RB, Liberles SD, et al. , Liraglutide modulates appetite and body weight through glucagon-like peptide 1 receptor-expressing glutamatergic neurons, Diabetes 67 (2018) 1538–1548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Egecioglu E, Engel JA, Jerlhag E, The glucagon-like peptide 1 analogue exendin-4 attenuates the nicotine-induced locomotor stimulation, accumbal dopamine release, conditioned place preference as well as the expression of locomotor sensitization in mice, PLoS ONE (2013) 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Perkins KA, Scott J, Sex differences in long-term smoking cessation rates due to nicotine patch, Nicotine Tob. Res 10 (2008) 1245–1251. [DOI] [PubMed] [Google Scholar]
  • [89].Perkins KA, Smoking cessation in women. Special considerations, CNS Drugs 15 (2001) 391–411. [DOI] [PubMed] [Google Scholar]
  • [90].Wolfman SL, Gill DF, Bogdanic F, Long K, Al-Hasani R, McCall JG, et al. , Nicotine aversion is mediated by GABAergic interpeduncular nucleus inputs to laterodorsal tegmentum, Nat. Commun 9 (2018) 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Antolin-Fontes B, Ables JL, Görlich A, Ibañez-Tallon I, The Habenulo-Interpeduncular Pathway in Nicotine Aversion and Withdrawal, Neuropharmacology: Elsevier Ltd, 2015, pp. 213–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Fowler CD, Kenny PJ, Nicotine aversion: neurobiological mechanisms and relevance to tobacco dependence vulnerability, Neuropharmacology 76 (2014) 533–544. Pt B. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Frahm S, Ślimak MA, Ferrarese L, Santos-Torres J, Antolin-Fontes B, Auer S, et al. , Aversion to nicotine is regulated by the balanced activity of β4 and α5 nicotinic receptor subunits in the medial habenula, Neuron 70 (2011) 522–535. [DOI] [PubMed] [Google Scholar]
  • [94].Fowler CD, Lu Q, Johnson PM, Marks MJ, Kenny PJ, Habenular α5 nicotinic receptor subunit signalling controls nicotine intake, Nature 471 (2011) 597–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Falk S, Petersen J, Svendsen C, Romero-Leguizamón CR, Jørgensen SH, Krauth N, et al. , GLP-1 and nicotine combination therapy engages hypothalamic and mesolimbic pathways to reverse obesity, Cell Rep (2023) 112466. [DOI] [PubMed] [Google Scholar]
  • [96].Hernandez NS, Ige KY, Mietlicki-Baase EG, Molina-Castro GC, Turner CA, Hayes MR, et al. , Glucagon-like peptide-1 receptor activation in the ventral tegmental area attenuates cocaine seeking in rats, Neuropsychopharmacology 43 (2018) 2000–2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Hernandez NS, O’Donovan B, Ortinski PI, Schmidt HD, Activation of glucagon-like peptide-1 receptors in the nucleus accumbens attenuates cocaine seeking in rats, Addict. Biol 24 (2019) 170–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Schmidt HD, Mietlicki-Baase EG, Ige KY, Maurer JJ, Reiner DJ, Zimmer DJ, et al. , Glucagon-like peptide-1 receptor activation in the ventral tegmental area decreases the reinforcing efficacy of cocaine, Neuropsychopharmacology 41 (2016) 1917–1928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Fortin SM, Roitman MF, Central GLP-1 receptor activation modulates cocaine-evoked phasic dopamine signaling in the nucleus accumbens core, Physiol. Behav 176 (2017) 17–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Gu G, Roland B, Tomaselli K, Dolman CS, Lowe C, Heilig JS, Glucagon-like peptide-1 in the rat brain: distribution of expression and functional implication, J. Comp. Neurol 521 (2013) 2235–2261. [DOI] [PubMed] [Google Scholar]
  • [101].Zhu C, Hong T, Li H, Jiang S, Guo B, Wang L, et al. , Glucagon-like peptide-1 agonist exendin-4 facilitates the extinction of cocaine-induced condition place preference, Front. Syst. Neurosci 15 (2021) 711750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Millan EZ, Marchant NJ, McNally GP, Extinction of drug seeking, Behav. Brain Res 217 (2011) 454–462. [DOI] [PubMed] [Google Scholar]
  • [103].Mietlicki-Baase EG, Ortinski PI, Rupprecht LE, Olivos DR, Alhadeff AL, Christopher Pierce R, et al. , The food intake-suppressive effects of glucagon-like peptide-1 receptor signaling in the ventral tegmental area are mediated by AMPA/kainate receptors, Am. J. Physiol. - Endocrinol. Metabol 305 (2013) 1367–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Liu J, Conde K, Zhang P, Lilascharoen V, Xu Z, Lim BK, et al. , Enhanced AMPA receptor trafficking mediates the anorexigenic effect of endogenous glucagon-like peptide-1 in the paraventricular hypothalamus, Neuron 96 (2017) 897–909, e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Ohtake N, Saito M, Eto M, Seki K, Exendin-4 promotes the membrane trafficking of the AMPA receptor GluR1 subunit and ADAM10 in the mouse neocortex, Regul. Pept 190–191 (2014) 1–11. [DOI] [PubMed] [Google Scholar]
  • [106].Park SW, Mansur RB, Lee Y, Lee JH, Seo MK, Choi AJ, et al. , Liraglutide activates mTORC1 signaling and AMPA receptors in rat hippocampal neurons under toxic conditions, Front. Neurosci 12 (2018) 756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Chesworth R, Corbit LH, Recent developments in the behavioural and pharmacological enhancement of extinction of drug seeking, Addict. Biol 22 (2017) 3–43. [DOI] [PubMed] [Google Scholar]
  • [108].Sutton MA, Schmidt EF, Choi K-H, Schad CA, Whisler K, Simmons D, et al. , Extinction-induced upregulation in AMPA receptors reduces cocaine-seeking behaviour, Nature 421 (2003) 70–75. [DOI] [PubMed] [Google Scholar]
  • [109].Epstein DH, Preston KL, Stewart J, Shaham Y, Toward a model of drug relapse: an assessment of the validity of the reinstatement procedure, Psychopharmacology (Berl.) (2006) 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Schmidt HD, Rupprecht LE, A Addy N, Neurobiological and neurophysiological mechanisms underlying nicotine seeking and smoking relapse, Mol. Neuropsychiatry 4 (2018) 169–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Hays JT, Ebbert JO, Sood A, Efficacy and safety of varenicline for smoking cessation, Am. J. Med 121 (2008) S32–S42. [DOI] [PubMed] [Google Scholar]
  • [112].O’Connor EC, Parker D, Rollema H, Mead AN, The alpha4beta2 nicotinic acetylcholine-receptor partial agonist varenicline inhibits both nicotine self-administration following repeated dosing and reinstatement of nicotine seeking in rats, Psychopharmacology (Berl) 208 (2010) 365–376. [DOI] [PubMed] [Google Scholar]
  • [113].Le Foll B, Chakraborty-Chatterjee M, Lev-Ran S, Barnes C, Pushparaj A, Gamaleddin I, et al. , Varenicline decreases nicotine self-administration and cue-induced reinstatement of nicotine-seeking behaviour in rats when a long pretreatment time is used, Int. J. Neuropsychopharmacol 15 (2012) 1265–1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Farr OM, Sofopoulos M, Tsoukas MA, Dincer F, Thakkar B, Sahin-Efe A, et al. , GLP-1 receptors exist in the parietal cortex, hypothalamus and medulla of human brains and the GLP-1 analogue liraglutide alters brain activity related to highly desirable food cues in individuals with diabetes: a crossover, randomised, placebo-controlled trial, Diabetologia 59 (2016) 954–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Klausen MK, Jensen ME, Møller M, Le Dous N, Jensen A, Zeeman VA, et al. , Exenatide once weekly for alcohol use disorder investigated in a randomized, placebo-controlled clinical trial, JCI Insight (2022) 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Konanur VR, Hsu TM, Kanoski SE, Hayes MR, Roitman MF, Phasic dopamine responses to a food-predictive cue are suppressed by the glucagon-like peptide-1 receptor agonist Exendin-4, Physiol. Behav 215 (2020) 112771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Liu X, Jernigen C, Gharib M, Booth S, Caggiula AR, Sved AF, Effects of dopamine antagonists on drug cue-induced reinstatement of nicotine-seeking behavior in rats, Behav. Pharmacol 21 (2010) 153–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Gipson CD, Reissner KJ, Kupchik YM, Smith AC, Stankeviciute N, Hensley-Simon ME, et al. , Reinstatement of nicotine seeking is mediated by glutamatergic plasticity, Proc. Natl. Acad. Sci. U. S. A 110 (2013) 9124–9129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Hamid S, Dawe GS, Gray JA, Stephenson JD, Nicotine induces long-lasting potentiation in the dentate gyrus of nicotine-primed rats, Neurosci. Res 29 (1997) 81–85. [DOI] [PubMed] [Google Scholar]
  • [120].Zhang TA, Tang J, Pidoplichko VI, Dani JA, Addictive nicotine alters local circuit inhibition during the induction of in vivo hippocampal synaptic potentiation, J. Neurosci 30 (2010) 6443–6453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Rebosio C, Balbi M, Passalacqua M, Ricciarelli R, Fedele E, Presynaptic GLP-1 receptors enhance the depolarization-evoked release of glutamate and GABA in the mouse cortex and hippocampus, Biofactors 44 (2018) 148–157. [DOI] [PubMed] [Google Scholar]
  • [122].Ratner RE, Maggs D, Nielsen LL, Stonehouse AH, Poon T, Zhang B, et al. , Long-term effects of exenatide therapy over 82 weeks on glycaemic control and weight in over-weight metformin-treated patients with type 2 diabetes mellitus, Diabetes Obes. Metab 8 (2006) 419–428. [DOI] [PubMed] [Google Scholar]
  • [123].Bettge K, Kahle M, Abd El Aziz MS, Meier JJ, A Nauck M, Occurrence of nausea, vomiting and diarrhoea reported as adverse events in clinical trials studying glucagon-like peptide-1 receptor agonists: a systematic analysis of published clinical trials, Diabetes, Obes. Metab 19 (2017) 336–347. [DOI] [PubMed] [Google Scholar]
  • [124].Vilsbøll T, Christensen M, Junker AE, Knop FK, Gluud LL, Effects of glucagon-like peptide-1 receptor agonists on weight loss: systematic review and meta-analyses of randomised controlled trials, BMJ 344 (2012) d7771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Mitchell D, Wells C, Hoch N, Lind K, Woods SC, Mitchell LK, Poison induced pica in rats, Physiol. Behav 17 (1976) 691–697. [DOI] [PubMed] [Google Scholar]
  • [126].Kanoski SE, Rupprecht LE, Fortin SM, De Jonghe BC, Hayes MR, The role of nausea in food intake and body weight suppression by peripheral GLP-1 receptor agonists, exendin-4 and liraglutide, Neuropharmacology 62 (2012) 1916–1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Babic I, Gorak A, Engel M, Sellers D, Else P, Osborne AL, et al. , Liraglutide prevents metabolic side-effects and improves recognition and working memory during antipsychotic treatment in rats, J. Psychopharmacol 32 (2018) 578–590. [DOI] [PubMed] [Google Scholar]
  • [128].Yammine L, Green CE, Kosten TR, de Dios C, Suchting R, Lane SD, et al. , Exenatide adjunct to nicotine patch facilitates smoking cessation and may reduce post-cessation weight gain: a pilot randomized controlled trial, Nicotine Tob. Res 23 (2021) 1682–1690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Lengsfeld S, Burkard T, Meienberg A, Jeanloz N, Vukajlovic T, Bologna K, et al. , Effect of dulaglutide in promoting abstinence during smoking cessation: a single-centre, randomized, double-blind, placebo-controlled, parallel group trial, EClinicalMedicine 57 (2023) 101865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Guo X, Zhou Z, Lyu X, Xu H, Zhu H, Pan H, et al. , The antiobesity effect and safety of GLP-1 receptor agonist in overweight/obese patients without diabetes: a systematic review and meta-analysis, Horm. Metab. Res 54 (2022) 458–471. [DOI] [PubMed] [Google Scholar]
  • [131].McLaughlin I, Dani JA, De Biasi M, Nicotine withdrawal, Curr. Top. Behav. Neurosci 24 (2015) 99–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Heishman SJ, Kleykamp BA, Singleton EG, Meta-analysis of the acute effects of nicotine and smoking on human performance, Psychopharmacology (Berl) 210 (2010) 453–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Orleans CT, Rimer BK, Cristinzio S, Keintz MK, Fleisher L, A national survey of older smokers: treatment needs of a growing population, Health Psychol 10 (1991) 343–351. [DOI] [PubMed] [Google Scholar]
  • [134].McKee SA, O’Malley SS, Salovey P, Krishnan-Sarin S, Mazure CM, Perceived risks and benefits of smoking cessation: gender-specific predictors of motivation and treatment outcome, Addict. Behav 30 (2005) 423–435. [DOI] [PubMed] [Google Scholar]
  • [135].Robinson JD, Li L, Chen M, Lerman C, Tyndale RF, Schnoll RA, et al. , Evaluating the temporal relationships between withdrawal symptoms and smoking relapse, Psychol. Addict. Behav 33 (2019) 105–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Malin DH, Lake JR, Carter VA, Cunningham JS, Hebert KM, Conrad DL, et al. , The nicotinic antagonist mecamylamine precipitates nicotine abstinence syndrome in the rat, Psychopharmacology (Berl) 115 (1994) 180–184. [DOI] [PubMed] [Google Scholar]
  • [137].De Biasi M, Salas R, Influence of neuronal nicotinic receptors over nicotine addiction and withdrawal, Exp. Biol. Med. (Maywood) 233 (2008) 917–929. [DOI] [PubMed] [Google Scholar]
  • [138].Rennard SI, Daughton DM, Smoking cessation, Clin. Chest Med 35 (2014) 165–176. [DOI] [PubMed] [Google Scholar]
  • [139].Kruger J, Ham SA, Prohaska TR, Behavioral risk factors associated with overweight and obesity among older adults: the 2005 National Health Interview Survey, Prev. Chronic Dis 6 (2009) A14. [PMC free article] [PubMed] [Google Scholar]
  • [140].Lycett D, Munafó M, Johnstone E, Murphy M, Aveyard P, Associations between weight change over 8 years and baseline body mass index in a cohort of continuing and quitting smokers, Addiction 106 (2011) 188–196. [DOI] [PubMed] [Google Scholar]
  • [141].O’Hara P, Connett JE, Lee WW, Nides M, Murray R, Wise R, Early and late weight gain following smoking cessation in the lung health study, Am. J. Epidemiol 148 (1998) 821–830. [DOI] [PubMed] [Google Scholar]
  • [142].Filozof C, Fernández Pinilla MC, Fernández-Cruz A, Smoking cessation and weight gain, Obes. Rev 5 (2004) 95–103. [DOI] [PubMed] [Google Scholar]
  • [143].Klesges RC, Winders SE, Meyers AW, Eck LH, Ward KD, Hultquist CM, et al. , How much weight gain occurs following smoking cessation? A comparison of weight gain using both continuous and point prevalence abstinence, J. Consult. Clin. Psychol 65 (1997) 286–291. [DOI] [PubMed] [Google Scholar]
  • [144].Swan GE, Ward MM, Carm Elli D, Jack LM, Differential rates of relapse in subgroups of male and female smokers, J. Clin. Epidemiol 46 (1993) 1041–1053. [DOI] [PubMed] [Google Scholar]
  • [145].Anker JJ, Nakajima M, Raatz S, Allen S, Absi M, Tobacco withdrawal increases junk food intake: the role of the endogenous opioid system, Drug Alcohol Depend 225 (2021) 108819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Takeda K, Aotani D, Kuga Y, Jinno T, Guo T, Ogawa K, et al. , A mouse model of weight gain after nicotine withdrawal, Biochem. Biophys. Res. Commun 588 (2022) 140–146. [DOI] [PubMed] [Google Scholar]
  • [147].Bellinger LL, Wellman PJ, Cepeda-Benito A, Kramer PR, Guan G, Tillberg CM, et al. , Meal patterns in female rats during and after intermittent nicotine administration, Pharmacol. Biochem. Behav 80 (2005) 437–444. [DOI] [PubMed] [Google Scholar]
  • [148].Grunberg NE, Bowen DJ, Winders SE, Effects of nicotine on body weight and food consumption in female rats, Psychopharmacology (Berl) 90 (1986) 101–105. [DOI] [PubMed] [Google Scholar]
  • [149].Grunberg NE, Bowen DJ, Morse DE, Effects of nicotine on body weight and food consumption in rats, Psychopharmacology (Berl) 83 (1984) 93–98. [DOI] [PubMed] [Google Scholar]
  • [150].Bláha V, Yang ZJ, Meguid M, Chai JK, Zadák Z, Systemic nicotine administration suppresses food intake via reduced meal sizes in both male and female rats, Acta Medica (Hradec. Kralove) 41 (1998) 167–173. [PubMed] [Google Scholar]
  • [151].McPherson RA, Pincus MR, Henry’s Clinical Diagnosis and Management By Laboratory Methods E-book, Elsevier Health Sciences, 2021. [Google Scholar]
  • [152].Lüthi H, Lengsfeld S, Burkard T, Meienberg A, Jeanloz N, Vukajlovic T, et al. , Effect of dulaglutide in promoting abstinence during smoking cessation: 12-month follow-up of a single-centre, randomised, double-blind, placebo-controlled, parallel group trial, eClinicalMedicine 68 (2024) 102429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Wilding JP, Batterham RL, Davies M, Van Gaal LF, Kandler K, Konakli K, et al. , Weight regain and cardiometabolic effects after withdrawal of semaglutide: the STEP 1 trial extension, Diabetes, Obes. Metabol 24 (2022) 1553–1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Rubino D, Abrahamsson N, Davies M, Hesse D, Greenway FL, Jensen C, et al. , Effect of continued weekly subcutaneous semaglutide vs placebo on weight loss maintenance in adults with overweight or obesity: the STEP 4 randomized clinical trial, JAMA 325 (2021) 1414–1425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Miyata G, Meguid MM, Fetissov SO, Torelli GF, Kim H-J, Nicotine’s effect on hypothalamic neurotransmitters and appetite regulation, Surgery 126 (1999) 255–263. [PubMed] [Google Scholar]
  • [156].Miyata G, Meguid MM, Varma M, Fetissov SO, Kim HJ, Nicotine alters the usual reciprocity between meal size and meal number in female rat, Physiol. Behav 74 (2001) 169–176. [DOI] [PubMed] [Google Scholar]
  • [157].Bellinger L, Cepeda-Benito A, Wellman PJ, Meal patterns in male rats during and after intermittent nicotine administration, Pharmacol. Biochem. Behav 74 (2003) 495–504. [DOI] [PubMed] [Google Scholar]
  • [158].Epstein LH, Wright SM, Paluch RA, Leddy J, Hawk LW Jr., Jaroni JL, et al. , Food hedonics and reinforcement as determinants of laboratory food intake in smokers, Physiol. Behav 81 (2004) 511–517. [DOI] [PubMed] [Google Scholar]
  • [159].Jias LM, Ellison G, Chronic nicotine induces a specific appetite for sucrose in rats, Pharmacol. Biochem. Behav 35 (1990) 489–491. [DOI] [PubMed] [Google Scholar]
  • [160].Mannucci C, Catania MA, Adamo EB, Bellomo M, Caputi AP, Calapai G, Long-term effects of high doses of nicotine on feeding behavior and brain nitric oxide synthase activity in female mice, J. Pharmacol. Sci 98 (2005) 232–238. [DOI] [PubMed] [Google Scholar]
  • [161].Caggiula AR, Donny EC, White AR, Chaudhri N, Booth S, Gharib MA, et al. , Cue dependency of nicotine self-administration and smoking, Pharmacol. Biochem. Behav 70 (2001) 515–530. [DOI] [PubMed] [Google Scholar]
  • [162].Donny EC, Caggiula AR, Weaver MT, Levin ME, Sved AF, The reinforcement-enhancing effects of nicotine: implications for the relationship between smoking, eating and weight, Physiol. Behav 104 (2011) 143–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Volkow ND, Wang G-J, Fowler JS, Telang F, Overlapping neuronal circuits in addiction and obesity: evidence of systems pathology, Philos. Trans. R. Soc. B: Biol. Sci 363 (2008) 3191–3200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Epping-Jordan MP, Watkins SS, Koob GF, Markou A, Dramatic decreases in brain reward function during nicotine withdrawal, Nature 393 (1998) 76–79. [DOI] [PubMed] [Google Scholar]
  • [165].Zhang L, Dong Y, Doyon WM, Dani JA, Withdrawal from chronic nicotine exposure alters dopamine signaling dynamics in the nucleus accumbens, Biol. Psychiatry 71 (2012) 184–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Guy EG, Choi E, Pratt WE, Nucleus accumbens dopamine and mu-opioid receptors modulate the reinstatement of food-seeking behavior by food-associated cues, Behav. Brain Res 219 (2011) 265–272. [DOI] [PubMed] [Google Scholar]
  • [167].Heinrichs SC, Lapsansky J, Behan DP, Chan RKW, Sawchenko PE, Lorang M, et al. , Corticotropin-releasing factor-binding protein ligand inhibitor blunts excessive weight gain in genetically obese Zucker rats and rats during nicotine withdrawal, Proc. Natl. Acad. Sci 93 (1996) 15475–15480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Dandekar MP, Nakhate KT, Kokare DM, Subhedar NK, Effect of nicotine on feeding and body weight in rats: involvement of cocaine- and amphetamine-regulated transcript peptide, Behav. Brain Res 219 (2011) 31–38. [DOI] [PubMed] [Google Scholar]
  • [169].Geloneze B, de Lima-Júnior JC, Velloso LA, Glucagon-like peptide-1 receptor agonists (GLP-1RAs) in the brain–adipocyte axis, Drugs 77 (2017) 493–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Ghosal S, Myers B, Herman JP, Role of central glucagon-like peptide-1 in stress regulation, Physiol. Behav 122 (2013) 201–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Harris KK, Zopey M, Friedman TC, Metabolic effects of smoking cessation, Nat. Rev. Endocrinol 12 (2016) 299–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Jacobsen LK, Krystal JH, Mencl WE, Westerveld M, Frost SJ, Pugh KR, Effects of smoking and smoking abstinence on cognition in adolescent tobacco smokers, Biol. Psychiatry 57 (2005) 56–66. [DOI] [PubMed] [Google Scholar]
  • [173].Cole DM, Beckmann CF, Long CJ, Matthews PM, Durcan MJ, Beaver JD, Nicotine replacement in abstinent smokers improves cognitive withdrawal symptoms with modulation of resting brain network dynamics, Neuroimage 52 (2010) 590–599. [DOI] [PubMed] [Google Scholar]
  • [174].Patterson F, Jepson C, Loughead J, Perkins K, Strasser AA, Siegel S, et al. , Working memory deficits predict short-term smoking resumption following brief abstinence, Drug Alcohol Depend 106 (2010) 61–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Hare TA, Camerer CF, Rangel A, Self-control in decision-making involves modulation of the vmPFC valuation system, Science 324 (2009) 646–648. [DOI] [PubMed] [Google Scholar]
  • [176].Kouneiher F, Charron S, Koechlin E, Motivation and cognitive control in the human prefrontal cortex, Nat. Neurosci 12 (2009) 939–945. [DOI] [PubMed] [Google Scholar]
  • [177].Ashare RL, Schmidt HD, Optimizing treatments for nicotine dependence by increasing cognitive performance during withdrawal, Expert Opin. Drug Discov 9 (2014) 579–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Ashare RL, Falcone M, Lerman C, Cognitive function during nicotine withdrawal: implications for nicotine dependence treatment, Neuropharmacology (2014) 76. Pt B:581–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Lerman C, LeSage MG, Perkins KA, O’Malley SS, Siegel SJ, Benowitz NL, et al. , Translational research in medication development for nicotine dependence, Nat. Rev. Drug Discov 6 (2007) 746–762. [DOI] [PubMed] [Google Scholar]
  • [180].Birks J, Cholinesterase inhibitors for Alzheimer’s disease, Cochrane Database Syst. Rev (2006), 2006:Cd005593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Wilkinson DS, Gould TJ, The effects of galantamine on nicotine withdrawal-induced deficits in contextual fear conditioning in C57BL/6 mice, Behav. Brain Res 223 (2011) 53–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Kimmey BA, Rupprecht LE, Hayes MR, Schmidt HD, Donepezil, an acetylcholinesterase inhibitor, attenuates nicotine self-administration and reinstatement of nicotine seeking in rats, Addict. Biol 19 (2014) 539–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Hopkins TJ, Rupprecht LE, Hayes MR, Blendy JA, Schmidt HD, Galantamine, an acetylcholinesterase inhibitor and positive allosteric modulator of nicotinic acetylcholine receptors, attenuates nicotine taking and seeking in rats, Neuropsychopharmacology 37 (2012) 2310–2321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Ashare RL, Kimmey BA, Rupprecht LE, Bowers ME, Hayes MR, Schmidt HD, Repeated administration of an acetylcholinesterase inhibitor attenuates nicotine taking in rats and smoking behavior in human smokers, Transl. Psychiatry 6 (2016) e713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Patterson F, Jepson C, Strasser AA, Loughead J, Perkins KA, Gur RC, et al. , Varenicline improves mood and cognition during smoking abstinence, Biol. Psychiatry 65 (2009) 144–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Ashare RL, McKee SA, Effects of varenicline and bupropion on cognitive processes among nicotine-deprived smokers, Exp. Clin. Psychopharmacol 20 (2012) 63–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Cahill K, Stead LF, Lancaster T, Nicotine receptor partial agonists for smoking cessation, Cochrane Database Syst. Rev (2011). Cd006103. [DOI] [PubMed] [Google Scholar]
  • [188].Hughes JR, Stead LF, Lancaster T, Antidepressants for smoking cessation, Cochrane Database Syst. Rev (2007). Cd000031. [DOI] [PubMed] [Google Scholar]
  • [189].Abd el-Rady NM, Ahmed A, Abdel-Rady MM, I Ismail O, Glucagon-like peptide-1 analog improves neuronal and behavioral impairment and promotes neuroprotection in a rat model of aluminum-induced dementia, Physiol. Rep 8 (2021) e14651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Palleria C, Leo A, Andreozzi F, Citraro R, Iannone M, Spiga R, et al. , Liraglutide prevents cognitive decline in a rat model of streptozotocin-induced diabetes independently from its peripheral metabolic effects, Behav. Brain Res 321 (2017) 157–169. [DOI] [PubMed] [Google Scholar]
  • [191].Zhang M, Wu Y, Gao R, Chen X, Chen R, Chen Z, Glucagon-like peptide-1 analogs mitigate neuroinflammation in Alzheimer’s disease by suppressing NLRP2 activation in astrocytes, Mol. Cell. Endocrinol 542 (2022) 111529. [DOI] [PubMed] [Google Scholar]
  • [192].Isacson R, Nielsen E, Dannaeus K, Bertilsson G, Patrone C, Zachrisson O, et al. , The glucagon-like peptide 1 receptor agonist exendin-4 improves reference memory performance and decreases immobility in the forced swim test, Eur. J. Pharmacol 650 (2011) 249–255. [DOI] [PubMed] [Google Scholar]
  • [193].Abdelwahed OM, Tork OM, Gamal el Din MM, Rashed L, M Zickri, Effect of glucagon-like peptide-1 analogue; Exendin-4, on cognitive functions in type 2 diabetes mellitus; possible modulation of brain derived neurotrophic factor and brain Visfatin, Brain Res. Bull 139 (2018) 67–80. [DOI] [PubMed] [Google Scholar]
  • [194].Zhang L-Q, Zhang W, Li T, Yang T, Yuan X, Zhou Y, et al. , GLP-1R activation ameliorated novel-object recognition memory dysfunction via regulating hippocampal AMPK/NF-κB pathway in neuropathic pain mice, Neurobiol. Learn. Mem 182 (2021) 107463. [DOI] [PubMed] [Google Scholar]
  • [195].Seo MK, Jeong S, Seog D-H, Lee JA, Lee J-H, Lee Y, et al. , Effects of liraglutide on depressive behavior in a mouse depression model and cognition in the probe trial of Morris water maze test, J. Affect. Disord 324 (2023) 8–15. [DOI] [PubMed] [Google Scholar]
  • [196].Kamble M, Gupta R, Rehan HS, Gupta LK, Neurobehavioral effects of liraglutide and sitagliptin in experimental models, Eur. J. Pharmacol 774 (2016) 64–70. [DOI] [PubMed] [Google Scholar]
  • [197].Pelle MC, Zaffina I, Giofrè F, Pujia R, Arturi F, Potential role of glucagon-like peptide-1 receptor agonists in the treatment of cognitive decline and dementia in diabetes mellitus, Int. J. Mol. Sci 24 (2023) 11301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Vadini F, Simeone PG, Boccatonda A, Guagnano MT, Liani R, Tripaldi R, et al. , Liraglutide improves memory in obese patients with prediabetes or early type 2 diabetes: a randomized, controlled study, Int. J. Obes 44 (2020) 1254–1263. [DOI] [PubMed] [Google Scholar]
  • [199].Li Q, Jia M, Yan Z, Li Q, Sun F, He C, et al. , Activation of glucagon-like peptide-1 receptor ameliorates cognitive decline in type 2 diabetes mellitus through a metabolism-independent pathway, J. Am. Heart Assoc 10 (2021) e020734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Cheng H, Zhang Z, Zhang B, Zhang W, Wang J, Ni W, et al. , Enhancement of impaired olfactory neural activation and cognitive capacity by liraglutide, but not dapagliflozin or acarbose, in patients with type 2 diabetes: a 16-week randomized parallel comparative study, Diabetes Care 45 (2022) 1201–1210. [DOI] [PubMed] [Google Scholar]
  • [201].Cukierman-Yaffe T, Gerstein HC, Colhoun HM, Diaz R, García-Pérez L-E, Lakshmanan M, et al. , Effect of dulaglutide on cognitive impairment in type 2 diabetes: an exploratory analysis of the REWIND trial, Lancet Neurol 19 (2020) 582–590. [DOI] [PubMed] [Google Scholar]
  • [202].Saravia R, Ten-Blanco M, Grande MT, Maldonado R, Berrendero F, Anti-inflammatory agents for smoking cessation? Focus on cognitive deficits associated with nicotine withdrawal in male mice, Brain Behav. Immun 75 (2019) 228–239. [DOI] [PubMed] [Google Scholar]
  • [203].Zilliox LA, Chadrasekaran K, Kwan JY, Russell JW, Diabetes and cognitive impairment, Curr. Diab. Rep 16 (2016) 87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Hughes JR, Effects of abstinence from tobacco: valid symptoms and time course, Nicotine Tob. Res 9 (2007) 315–327. [DOI] [PubMed] [Google Scholar]
  • [205].Crawley JN, Belknap JK, Collins A, Crabbe JC, Frankel W, Henderson N, et al. , Behavioral phenotypes of inbred mouse strains: implications and recommendations for molecular studies, Psychopharmacology (Berl) 132 (1997) 107–124. [DOI] [PubMed] [Google Scholar]
  • [206].Cryan JF, Markou A, Lucki I, Assessing antidepressant activity in rodents: recent developments and future needs, Trends Pharmacol. Sci 23 (2002) 238–245. [DOI] [PubMed] [Google Scholar]
  • [207].Kenny PJ, Markou A, Neurobiology of the nicotine withdrawal syndrome, Pharmacol. Biochem. Behav 70 (2001) 531–549. [DOI] [PubMed] [Google Scholar]
  • [208].Crawley J, Goodwin FK, Preliminary report of a simple animal behavior model for the anxiolytic effects of benzodiazepines, Pharmacol. Biochem. Behav 13 (1980) 167–170. [DOI] [PubMed] [Google Scholar]
  • [209].Stoker AK, Semenova S, Markou A, Affective and somatic aspects of spontaneous and precipitated nicotine withdrawal in C57BL/6J and BALB/cByJ mice, Neuropharmacology 54 (2008) 1223–1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Cryan JF, Mombereau C, Vassout A, The tail suspension test as a model for assessing antidepressant activity: review of pharmacological and genetic studies in mice, Neurosci. Biobehav. Rev 29 (2005) 571–625. [DOI] [PubMed] [Google Scholar]
  • [211].Schmidt HD, Duman RS, The role of neurotrophic factors in adult hippocampal neurogenesis, antidepressant treatments and animal models of depressive-like behavior, Behav. Pharmacol (2007) 18. [DOI] [PubMed] [Google Scholar]
  • [212].Thanos P, Delis F, Rosko L, Volkow ND, Passive response to stress in adolescent female and adult male mice after intermittent nicotine exposure in adolescence, J. Addict. Res. Ther (Suppl 6) (2013) 007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Sağlam C, İ Turan HS Özaçmak, The effect of glucagon like peptide-1 receptor agonist on behavioral despair and anxiety-like behavior in ovariectomized rats: modulation of BDNF/CREB, Nrf2 and lipocalin 2, Behav. Brain Res 435 (2022) 114053. [DOI] [PubMed] [Google Scholar]
  • [214].Darwish AB, El Sayed NS, Salama AAA, Saad MA, Dulaglutide impedes depressive-like behavior persuaded by chronic social defeat stress model in male C57BL/6 mice: implications on GLP-1R and cAMP/PKA signaling pathway in the hippocampus, Life Sci 320 (2023) 121546. [DOI] [PubMed] [Google Scholar]
  • [215].Ventorp F, Bay-Richter C, Nagendra AS, Janelidze S, Matsson VS, Lipton J, et al. , Exendin-4 treatment improves LPS-induced depressive-like behavior without affecting pro-inflammatory cytokines, J. Parkinsons Dis 7 (2017) 263–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Erbil D, Eren CY, Demirel C, Küçüker MU, Solaroğlu I, Y Eser H, GLP-1’s role in neuroprotection: a systematic review, Brain Inj 33 (2019) 734–819. [DOI] [PubMed] [Google Scholar]
  • [217].Gold PW, Goodwin FK, P Chrousos G, Clinical and biochemical manifestations of depression. Relation to the neurobiology of stress (2), N. Engl. J. Med 319 (1988) 413–420. [DOI] [PubMed] [Google Scholar]
  • [218].Zunszain PA, Anacker C, Cattaneo A, Carvalho LA, Pariante CM, Glucocorticoids, cytokines and brain abnormalities in depression, Prog. Neuropsychopharmacol. Biol. Psychiatry 35 (2011) 722–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Schmidt HD, Shelton RC, Duman RS, Functional biomarkers of depression: diagnosis, treatment, and pathophysiology, Neuropsychopharmacology 36 (2011) 2375–2394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Baron JA, Comi RJ, Cryns V, Brinck-Johnsen T, Mercer NG, The effect of cigarette smoking on adrenal cortical hormones, J. Pharmacol. Exp. Ther 272 (1995) 151–155. [PubMed] [Google Scholar]
  • [221].Field AE, Colditz GA, Willett WC, Longcope C, McKinlay JB, The relation of smoking, age, relative weight, and dietary intake to serum adrenal steroids, sex hormones, and sex hormone-binding globulin in middle-aged men, J. Clin. Endocrinol. Metab 79 (1994) 1310–1316. [DOI] [PubMed] [Google Scholar]
  • [222].Ussher M, West R, Evans P, Steptoe A, McEwen A, Clow A, et al. , Reduction in cortisol after smoking cessation among users of nicotine patches, Psychosom. Med 68 (2006) 299–306. [DOI] [PubMed] [Google Scholar]
  • [223].Semba J, Wakuta M, Maeda J, Suhara T, Nicotine withdrawal induces subsensitivity of hypothalamic-pituitary-adrenal axis to stress in rats: implications for precipitation of depression during smoking cessation, Psychoneuroendocrinology 29 (2004) 215–226. [DOI] [PubMed] [Google Scholar]
  • [224].Frederick SL, Reus VI, Ginsberg D, Hall SM, Munoz RF, Ellman G, Cortisol and response to dexamethasone as predictors of withdrawal distress and abstinence success in smokers, Biol. Psychiatry 43 (1998) 525–530. [DOI] [PubMed] [Google Scholar]
  • [225].Wong JA, Pickworth WB, Waters AJ, Absi M, Leventhal AM, Cortisol levels decrease after acute tobacco abstinence in regular smokers, Human Psychopharmacol 29 (2014) 152–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Absi M, Nakajima M, DeAngelis B, Grant J, King A, Grabowski J, et al. , Blunted opioid regulation of the HPA stress response during nicotine withdrawal: therapeutic implications, Stress 24 (2021) 529–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Gil-Lozano M, Pérez-Tilve D, Alvarez-Crespo M, Martís A, Fernandez AM, Catalina PAF, et al. , GLP-1(7–36)-amide and exendin-4 stimulate the HPA axis in rodents and humans, Endocrinology 151 (2010) 2629–2640. [DOI] [PubMed] [Google Scholar]
  • [228].Gil-Lozano M, Romaní-Pérez M, Outeiriño-Iglesias V, Vigo E, Brubaker PL, Gonźalez-Matías LC, et al. , Effects of prolonged exendin-4 administration on hypothalamic-pituitary-adrenal axis activity and water balance, Am. J. Physiol. Endocrinol. Metabol 304 (2013) E1105–E1E17. [DOI] [PubMed] [Google Scholar]
  • [229].Carlessi AS, Borba LA, Zugno AI, Quevedo J, Ŕeus GZ, Gut microbiota-brain axis in depression: the role of neuroinflammation, Eur. J. Neurosci 53 (2021) 222–235. [DOI] [PubMed] [Google Scholar]
  • [230].Setoyama D, Kato TA, Hashimoto R, Kunugi H, Hattori K, Hayakawa K, et al. , Plasma metabolites predict severity of depression and suicidal ideation in psychiatric patients-A multicenter pilot analysis, PLoS ONE 11 (2016) e0165267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Bansal Y, Kuhad A, Mitochondrial dysfunction in depression, Curr. Neuropharmacol 14 (2016) 610–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [232].Biedermann L, Zeitz J, Mwinyi J, Sutter-Minder E, Rehman A, Ott SJ, et al. , Smoking cessation induces profound changes in the composition of the intestinal microbiota in humans, PLoS ONE 8 (2013) e59260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Malińska D, Więckowski MR, Michalska B, Drabik K, Prill M, Patalas-Krawczyk P, et al. , Mitochondria as a possible target for nicotine action, J. Bioenerg. Biomembr 51 (2019) 259–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Moreira GV, Azevedo FF, Ribeiro LM, Santos A, Guadagnini D, Gama P, et al. , Liraglutide modulates gut microbiota and reduces NAFLD in obese mice, J. Nutr. Biochem 62 (2018) 143–154. [DOI] [PubMed] [Google Scholar]
  • [235].He W, Wang H, Zhao C, Tian X, Li L, Wang H, Role of liraglutide in brain repair promotion through Sirt1-mediated mitochondrial improvement in stroke, J. Cell Physiol 235 (2020) 2986–3001. [DOI] [PubMed] [Google Scholar]
  • [236].Kang MY, Oh TJ, Cho YM, Glucagon-like peptide-1 increases mitochondrial biogenesis and function in INS-1 rat insulinoma cells, Endocrinol. Metab. (Seoul) 30 (2015) 216–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Semenkovich K, Brown ME, Svrakic DM, Lustman PJ, Depression in type 2 diabetes mellitus: prevalence, impact, and treatment, Drugs 75 (2015) 577–587. [DOI] [PubMed] [Google Scholar]
  • [238].Milaneschi Y, Simmons WK, van Rossum EFC, Penninx BW, Depression and obesity: evidence of shared biological mechanisms, Mol. Psychiatry 24 (2019) 18–33. [DOI] [PubMed] [Google Scholar]
  • [239].Tsai W-H, Sung F-C, Chiu L-T, Shih Y-H, Tsai M-C, Wu S-I, Decreased risk of anxiety in diabetic patients receiving glucagon-like peptide-1 receptor agonist: a nationwide, population-based cohort study, Front. Pharmacol (2022) 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Wium-Andersen IK, Osler M, Jørgensen MB, Rungby J, Wium-Andersen MK, Diabetes, antidiabetic medications and risk of depression – a population-based cohort and nested case-control study, Psychoneuroendocrinology 140 (2022) 105715. [DOI] [PubMed] [Google Scholar]
  • [241].Pozzi M, Mazhar F, Peeters G, Vantaggiato C, Nobile M, Clementi E, et al. , A systematic review of the antidepressant effects of glucagon-like peptide 1 (GLP-1) functional agonists: further link between metabolism and psychopathology: special section on “Translational and Neuroscience Studies in Affective Disorders”. Section Editor, Maria Nobile MD, PhD. This section of JAD focuses on the relevance of translational and neuroscience studies in providing a better understanding of the neural basis of affective disorders. The main aim is to briefly summaries relevant research findings in clinical neuroscience with particular regards to specific innovative topics in mood and anxiety disorders, J. Affect. Disord (2019) 257. [DOI] [PubMed] [Google Scholar]
  • [242].Gamble JM, Chibrikov E, Midodzi WK, Twells LK, Majumdar SR, Examining the risk of depression or self-harm associated with incretin-based therapies used to manage hyperglycaemia in patients with type 2 diabetes: a cohort study using the UK Clinical Practice Research Datalink, BMJ Open 8 (2018) e023830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Kessing LV, Rytgaard HC, Ekstrøm CT, Knop FK, Berk M, Gerds TA, Antidiabetes agents and incident depression: a nationwide population-based study, Diabetes Care 43 (2020) 3050–3060. [DOI] [PubMed] [Google Scholar]
  • [244].Wang W, Volkow ND, Berger NA, Davis PB, Kaelber DC, Xu R, Association of semaglutide with risk of suicidal ideation in a real-world cohort, Nat. Med 30 (2024) 168–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [245].Cooper DH, Ramachandra R, Ceban F, Di Vincenzo JD, Rhee TG, Mansur RB, et al. , Glucagon-like peptide 1 (GLP-1) receptor agonists as a protective factor for incident depression in patients with diabetes mellitus: a systematic review, J. Psychiatr. Res 164 (2023) 80–89. [DOI] [PubMed] [Google Scholar]
  • [246].Kannel WB, Update on the role of cigarette smoking in coronary artery disease, Am. Heart J 101 (1981) 319–328. [DOI] [PubMed] [Google Scholar]
  • [247].Jeong S-M, Jeon KH, Shin DW, Han K, Kim D, Park SH, et al. , Smoking cessation, but not reduction, reduces cardiovascular disease incidence, Eur. Heart J 42 (2021) 4141–4153. [DOI] [PubMed] [Google Scholar]
  • [248].Bernaards CM, Twisk JWR, Snel J, van Mechelen W, Kemper HCG, In a prospective study in young people, associations between changes in smoking behavior and risk factors for cardiovascular disease were complex, J. Clin. Epidemiol 58 (2005) 1165–1171. [DOI] [PubMed] [Google Scholar]
  • [249].Duncan MS, Freiberg MS, Greevy RA Jr., Kundu S, Vasan RS, Tindle HA, Association of smoking cessation with subsequent risk of cardiovascular disease, JAMA 322 (2019) 642–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [250].Komiyama M, Wada H, Ura S, Yamakage H, Satoh-Asahara N, Shimada S, et al. , The effects of weight gain after smoking cessation on atherogenic α1-antitrypsin-low-density lipoprotein, Heart Vessels 30 (2015) 734–739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Hu Y, Zong G, Liu G, Wang M, Rosner B, Pan A, et al. , Smoking cessation, weight change, type 2 diabetes, and mortality, N. Engl. J. Med 379 (2018) 623–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Marks JB, Raskin P, Cardiovascular risk in diabetes: a brief review, J. Diabetes Complicat 14 (2000) 108–115. [DOI] [PubMed] [Google Scholar]
  • [253].Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, et al. , Liraglutide and cardiovascular outcomes in type 2 diabetes, N. Engl. J. Med 375 (2016) 311–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [254].Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, et al. , Semaglutide and cardiovascular outcomes in patients with type 2 diabetes, N. Engl. J. Med 375 (2016) 1834–1844. [DOI] [PubMed] [Google Scholar]
  • [255].Kosiborod MN, Bhatta M, Davies M, Deanfield JE, Garvey WT, Khalid U, et al. , Semaglutide improves cardiometabolic risk factors in adults with overweight or obesity: STEP 1 and 4 exploratory analyses, Diabetes Obes. Metab 25 (2023) 468–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [256].Rakipovski G, Rolin B, Nøhr J, Klewe I, Frederiksen KS, Augustin R, et al. , The GLP-1 analogs liraglutide and semaglutide reduce atherosclerosis in ApoE(−/−) and LDLr(−/−) mice by a mechanism that includes inflammatory pathways, JACC Basic Transl. Sci 3 (2018) 844–857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].McClernon FJ, Hiott FB, Huettel SA, Rose JE, Abstinence-induced changes in self-report craving correlate with event-related FMRI responses to smoking cues, Neuropsychopharmacology 30 (2005) 1940–1947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Yammine L, Kosten TR, Pimenova M, Schmitz JM, Cigarette smoking, type 2 diabetes mellitus, and glucagon-like peptide-1 receptor agonists as a potential treatment for smokers with diabetes: an integrative review, Diabetes Res. Clin. Pract 149 (2019) 78–88. [DOI] [PubMed] [Google Scholar]
  • [259].Iino K, Iwase M, Tsutsu N, Iida M, Smoking cessation and glycaemic control in type 2 diabetic patients, Diabetes Obes. Metab 6 (2004) 181–186. [DOI] [PubMed] [Google Scholar]
  • [260].Lycett D, Nichols L, Ryan R, Farley A, Roalfe A, Mohammed MA, et al. , The association between smoking cessation and glycaemic control in patients with type 2 diabetes: a THIN database cohort study, Lancet Diabetes Endocrinol 3 (2015) 423–430. [DOI] [PubMed] [Google Scholar]
  • [261].Alexander N, Rachel B, Nia R, Andrew F, Smoking cessation in adults with diabetes: a systematic review and meta-analysis of data from randomised controlled trials, BMJ Open 4 (2014) e004107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].Medaris A, A Woman Who Smoked Up to a Pack of Cigarettes a Day Found Them ‘Disgusting’ When She Started Taking Ozempic. Now She Hasn’t Smoked in 6 Months, Insider, 2023. [Google Scholar]
  • [263].Lu D, Can Diabetes and Weight-Loss Drug Ozempic Break Addictions Too? The Guardian, 2023. [Google Scholar]
  • [264].Calvo SS-C, Egan JM, The endocrinology of taste receptors, Nat. Rev. Endocrinol 11 (2015) 213–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [265].Martin B, Dotson CD, Shin Y-K, Ji S, Drucker DJ, Maudsley S, et al. , Modulation of taste sensitivity by GLP-1 signaling in taste buds, Ann. N. Y. Acad. Sci 1170 (2009) 98–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [266].Shin YK, Martin B, Golden E, Dotson CD, Maudsley S, Kim W, et al. , Modulation of taste sensitivity by GLP-1 signaling, J. Neurochem 106 (2008) 455–463. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No data was used for the research described in the article.

RESOURCES