Next Article in Journal
Prevalence of Chronic Obstructive Pulmonary Disease and Asthma in the Community of Pathumthani, Thailand
Previous Article in Journal
Ultrasound Relationship of Plantar Fat and Predislocation Syndrome
Previous Article in Special Issue
Electrolyte Imbalances and Metabolic Emergencies in Obesity: Mechanisms and Clinical Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current and Emerging Parenteral and Peroral Medications for Weight Loss: A Narrative Review

1
Sultan Qaboos University Hospital, Al-Khoud 123, P.O. Box 50, Muscat 123, Oman
2
Department of Medicine, Royal College of Surgeons, 123 St Stephen’s Green, D02 YN77 Dublin, Ireland
3
Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoudh 123, P.O. Box 50, Muscat 123, Oman
4
Pharmacy Program, Department of Pharmaceutics, Oman College of Health Sciences, P.O. Box 393, Muscat 113, Oman
*
Author to whom correspondence should be addressed.
Diseases 2025, 13(5), 129; https://doi.org/10.3390/diseases13050129
Submission received: 27 February 2025 / Revised: 3 April 2025 / Accepted: 8 April 2025 / Published: 22 April 2025

Abstract

:
Obesity is a growing global health challenge, necessitating effective treatment options beyond lifestyle interventions. This narrative review explores established and emerging pharmacotherapies for weight management, including parenteral agents like Liraglutide, Semaglutide, Setmelanotide, and Tirzepatide, as well as peroral medications such as Phentermine, Phentermine/Topiramate, Bupropion/Naltrexone, Orlistat, and Metformin. Newer treatments like Cagrilintide and Bimagrumab show promise for enhancing weight loss outcomes. Parenteral GLP-1 receptor agonists demonstrate superior efficacy compared to traditional peroral medications, with gastrointestinal side effects being the most common. Artificial intelligence presents intriguing opportunities to enhance weight loss strategies; however, its integration into clinical practice remains investigational and requires rigorous clinical validation. While current anti-obesity medications deliver significant benefits, future research must determine the efficacy, safety, and cost-effectiveness of AI-driven approaches. This includes exploring how AI can complement combination therapies and tailor personalized interventions, thereby grounding its potential benefits in robust clinical evidence. Future directions will focus on integrating AI into clinical trials to refine and personalize obesity management strategies.

1. Introduction

Obesity is a complex multifactorial disease that results in the collection of excess fat in the body, impairing overall health [1]. According to the World Health Organization (WHO), obesity is defined as a body mass index (BMI) ≥ 30 [2]. Obesity has been a growing concern over the past 50 years. In 1975, 100 million adults were living with obesity [3]. This number nearly quadrupled to 396 million by 2005, reached 609 million by 2015, and rose to 800 million in 2020, which increased to 890 million in 2022 [4,5,6,7]. The escalating burden of obesity is reflected in the disability-adjusted life years (DALYs) and mortality attributed to obesity. DALYs is a measurement of the overall disease burden and calculated by adding the number of years of life lost because of the disease to the years lived with disability [8,9]. Obesity-associated DALY rates were 160.3 million worldwide in 2019, making obesity responsible for 5.89% of global DALYs. In the same year, obesity was directly responsible for 8.52% of overall mortality, resulting in 5.03 million deaths. Compared to those in 1990, obesity-related DALYs and mortality nearly doubled, which highlights the increasing impact of obesity on global health [8,9].
Moreover, obesity is associated with multiple chronic diseases, including cardiovascular diseases (CVDs), type 2 diabetes (T2D), hypertension and dyslipidemia, with CVDs and T2D being the most strongly linked to obesity [10,11]. Interestingly, many of these comorbidities follow a dose-response relationship with obesity, meaning that the likelihood of being affected by them increases with higher body mass index (BMI) [12]. For example, one study shows that the likelihood of developing diabetes increased 17 times in individuals with a BMI ≥ 35 compared to those with a BMI between 25 and 29 [12]. In addition, a 1-unit gain in BMI can result in a 4% increase in the risk of atrial fibrillation [13], and a 5-unit increase in BMI can increase the risk of coronary heart disease by 29% [14]. Also, an additional 10 kg body weight is linked to a 3.0 mmHg increase in systolic blood pressure (SBP) and a 2.3 mmHg increase in diastolic blood pressure (DBP) [15].
Given the rising prevalence and health impact of obesity, effective management strategies are crucial. Current clinical guidelines for obesity management are based on the joint recommendations of the American Heart Association (AHA), American College of Cardiology (ACC), and The Obesity Society (TOS) from 2013, as well as the 2016 guidelines by the American Association of Clinical Endocrinologists (AACE) and American College of Endocrinology (ACE). AHA/ACC/TOS recommends a 5–10% reduction in the baseline weight in 6 months as an initial aim, which is enough to produce clinical benefits. A 5% weight reduction can considerably reduce the blood concentrations of substances associated with cardiometabolic diseases and enhance beta-cell function and insulin sensitivity of multiple organs, including the liver and adipose tissue [16,17]. Moreover, a 5–10% weight reduction has been shown to reduce SBP and DBP by 5 mmHg [18] and enhance metabolic diseases [16]. Lifestyle interventions are recommended for patients with BMI between 25 and 29.9 without obesity-related complications. The recommended daily calorie intake is 1200–1500 for females and 1500–1800 for males. Additionally, it is advised to engage in aerobic activity for 150–300 min at moderate intensity or 75–150 min at vigorous intensity per week, along with at least two strength exercise sessions. Pharmacological interventions are introduced to patients with a BMI ≥ 30 or a BMI ≥ 27 if they have obesity-associated comorbidity or if lifestyle interventions fail. Combining pharmacological and lifestyle interventions can lead to more significant weight reduction than lifestyle changes alone. Bariatric surgery is indicated for patients with a BMI ≥ 40 or a BMI ≥ 35 with obesity-associated complications [19,20,21].
Several reviews have examined current and emerging pharmacological approaches to weight loss; however, gaps remain in addressing other modalities such as lifestyle modifications and surgical procedures, as well as in directly comparing these strategies. Additionally, there is a need for more discussion on the role of artificial intelligence (AI) in obesity treatment, along with safety considerations and regulatory recommendations. This narrative review will address these gaps while exploring some emerging anti-obesity medications and both current and novel parenteral and peroral medications for weight loss with emphasis on their mechanisms of action, efficacy, and safety. The parenteral drugs in this review—such as Liraglutide, Semaglutide, Setmelanotide, and Tirzepatide—have gained attention due to their ability to modulate appetite and result and significant weight loss. The approach to treating obesity is also summarized in Figure 1.

2. Parenteral Medications for Weight Loss

Both surgical and pharmacological strategies have been developed to optimize therapeutic outcomes in managing chronic obesity. Among pharmacological treatments, parenteral drugs such as Liraglutide, Semaglutide, Setmelanotide, and Tirzepatide have promising potential.

2.1. Liraglutide (Saxenda)

Liraglutide, a glucagon-like peptide-1 (GLP1) receptor agonist, was initially developed for diabetes management. During clinical trials, it demonstrated potential in promoting weight reduction, leading to its approval for chronic weight management at a subcutaneous administration dose of 3 mg once daily. It is intended for long-term use, and studies have assessed its safety and efficacy over treatment periods of 1 to 2 years. Structurally, Liraglutide (Saxenda®) shares a 97% structural similarity with native human GLP-1 [22,23]. It includes a substitution of arginine for lysine at position 34 and a fatty acid side chain attached to lysine at position 26. It has a prolonged half-life of 10–14 h due to its additional albumin-binding fatty-acid side chain, which reduces its degradation by dipeptidyl peptidase-4 (DPP-4) and prolongs its half-life compared to endogenous GLP1 [24,25,26,27]. Over the treatment period of two years, Liraglutide was shown to reduce body weight by an average of 4.7%, primarily by activating GLP-1 receptors in the gastrointestinal and nervous systems. This interaction reduced energy intake, delayed gastric emptying, and increased satiety, contributing to weight loss [26,28]. Liraglutide crosses the blood-brain barrier through transcytosis, facilitated by tanycytes—specialized ependymoglial cells—and acts on hypothalamic GLP-1 receptors [29]. Once it is bound to the GLP-1 receptor, proopiomelanocortin and cocaine- and amphetamine-regulated transcript (POMC/CART) neurons are stimulated, and both neuropeptide Y (NPY) and agouti-related peptide (AgRP) are inhibited. This central mechanism helps regulate feelings of hunger and satiety, food intake, body weight, fat mass, and energy metabolism, including fatty acid oxidation [30]. Furthermore, Liraglutide has effectively reduced central adiposity, as indicated by decreased waist circumference and waist-to-hip ratio. It also significantly reduces android fat distribution.
Beyond weight loss, Liraglutide has demonstrated cardiovascular benefits, including lowering triglycerides (TRG) levels and improving the TRG/HDL ratio. In terms of the safety profile of Liraglutide, studies have reported a low risk of hypoglycemia in chronically obese, non-diabetic patients treated with 3 mg of Liraglutide, with an incidence of 1.3% compared to 1.0% in the placebo group [31]. Moreover, Liraglutide has shown promising therapeutic effects in obesity-related conditions, including obstructive sleep apnea. A clinical study demonstrated that 3 mg of Liraglutide significantly reduced the apnea–hypopnea index by 12.2 per hour, compared to 6.1 per hour in the placebo group. This improvement was due to the more significant weight reduction observed in the Liraglutide group (5.7%), compared to (1.6%) in the placebo group [32]. However, typical side effects of Liraglutide treatment include nausea (48.4%), vomiting (23.2%), and diarrhea (23.1%) [26,31,33,34,35,36]. Compared to other GLP-1 receptor agonists, Liraglutide has a shorter half-life, potentially leading to more frequent dosing and the return of hunger sensation between doses. In contrast, Semaglutide requires less frequent dosing with its longer half-life [37]. Notably, Liraglutide increases the risk of pancreatitis, with a reported incidence of 4%. Although this risk is relatively low, it is higher than other GLP-1 receptor agonists. As such, Liraglutide should be used with caution in patients at higher risk for pancreatitis (e.g., alcohol abuse, history of pancreatitis, cholelithiasis) [38,39].

2.2. Semaglutide (Wegovy)

Semaglutide, marketed under different brand names, was initially developed for treating type 2 diabetes (Ozempic®). However, recent studies have demonstrated its effectiveness in managing obesity, leading to the development of Wegovy® for this purpose. Clinical trials have shown promising therapeutic effects of Wegovy® in managing obesity, with evidence indicating that it can lead to a 15% reduction in body weight over 68 weeks [40,41]. Consequently, the U.S. Food and Drug Administration (FDA) has approved Semaglutide for chronic obesity at a dose of 2.4 mg once weekly. Semaglutide is a synthetic analogue of glucagon-like peptide 1 (GLP-1), sharing 94% amino acid homology with native GLP 1 [42]. GLP-1 is naturally synthesized by L-cells found in the small intestine and colon. Upon binding to GLP-1 receptors in tissues such as the brain, pancreas, and gut, it stimulates insulin secretion, inhibits glucagon release, slows gastric emptying, and enhances feelings of satiety, reducing food intake [43,44,45]. These combined actions are key to Semaglutide’s efficacy in weight reduction, as native GLP-1 has a short half-life of 2–11 min due to rapid degradation by the enzyme dipeptidyl peptidase-4 (DPP-4) [46]. To overcome the challenge of rapid degradation, Semaglutide was designed with a longer half-life of approximately one week, making it resistant to DPP-4 degradation and allowing for sustained therapeutic effects [42].
This extended half-life is achieved through two key modifications: the addition of a fatty diacid chain at position 26, which increases binding affinity to albumin, and the substitution of alanine with α-aminoisobutyric acid at position 8, making the molecule more resistant to DPP-4 degradation [41]. Additionally, evaluating the safety profile and side effects of Semaglutide is crucial for its clinical therapeutic use. Clinical trials have reported that Semaglutide is associated with gastrointestinal side effects such as nausea, vomiting, and diarrhea. In phase 3 trials, subcutaneous Semaglutide over 30 weeks induced nausea in 11.4 to 20% of patients (compared to 3.3–8% in the placebo group), vomiting in 4 to 11.5% (placebo 2–3%), and diarrhea in 4.5 to 11.3% (placebo 1.5–6%) [47,48,49]. Notably, these gastrointestinal side effects were reduced by 23% when dose escalation was carried out over 8 weeks instead of 4 weeks [50,51]. In terms of serious adverse events, a study observed acute gallbladder disease happening in approximately 4% of participants receiving once-weekly subcutaneous Semaglutide (2.4 mg). However, no cases of acute renal failure, diabetic retinopathy, or severe hypoglycemia were reported. Semaglutide has also been shown to significantly improve cardiometabolic risk factors, including reduced waist circumference and levels of glycated hemoglobin and lipids [52]. Notably, a meta-analysis of 37 studies found no significant association between Semaglutide and pancreatic or thyroid cancer. The risk of all neoplasms, including benign, malignant, and unspecified tumors, was similar between the Semaglutide and placebo groups, with no statistically significant differences observed [53].

2.3. Setmelanotide (Imcivree)

Setmelanotide (IMCIVREE®) is a melanocortin-4 (MC4) receptor agonist that was approved by the FDA in 2020 for the treatment of chronic genetic obesity [54,55]. It is designed for long-term use, and clinical trials have evaluated its safety and efficacy over treatment periods of one year or longer. The subcutaneous parenteral form of Setmelanotide is specifically indicated in patients with genetic defects in the MC4R activation pathway, such as variants in the proopiomelanocortin (POMC), proprotein convertase subtilisin/kexin type 1 (PCSK1), or leptin receptor (LEPR) genes. These defects impair the body’s natural weight regulation mechanisms, typically confirmed through genetic testing [54,55,56,57]. The MC4R activation pathway begins with leptin binding to LEPR, activating neurons that express POMC. POMC is cleaved by PCSK1, resulting in the production of ⍺-melanocyte-stimulating hormone (α-MSH), the endogenous ligand for the MC4 receptor. Activation of MC4 receptors plays a key role in regulating body weight [58]. Therefore, genetic deficiencies in any part of this pathway can lead to chronic obesity, which Setmelanotide can treat. Structurally, Setmelanotide is a cyclic peptide containing eight amino acids with a disulfide bond linking two cysteines near the termini. The molecule also features phenylalanine in the D-configuration within the conserved HFRW motif [59]. Setmelanotide’s effectiveness lies in its biased agonism, which strongly activates PLC-β (phospholipase C-β) via the G-protein Gαq. This unique mechanism allows Setmelanotide to resist antagonism by agouti-related peptide (AgRP), a natural antagonist of MC4R. As a result, Setmelanotide activates MC4R more effectively than the natural ligand α-MSH, leading to greater clinical benefits such as significant weight reduction [60]. Additionally, Setmelanotide has shown therapeutic efficacy in cases of chronic obesity caused by ciliopathic genetic syndromes, such as Bardet–Biedl syndrome (BBS), caused by a mutation in (BBS1-BBS22). In these cases, Setmelanotide bypasses the defective leptin pathway and directly activates the MC4 receptor, ultimately reducing weight [58].
As a result of the activation of the MC4 receptor by Setmelanotide, a variety of positive clinical therapeutic effects can be achieved, including reducing feelings of hunger, lowering calorie intake, and increasing energy expenditure, potentially leading to weight loss [61]. Clinical trials support Setmelanotide’s impact on weight loss. In trials involving patients with LEPR and POMC deficiencies (11 and 10 participants, respectively), 80% of participants in the POMC trial and 45% in the LEPR trial achieved 10% weight loss after one year. Changes in hunger feelings were reported as 27.1% (p = 0.0005) in the POMC trial and 43.7% (p < 0.0001) in the LEPR trial. Common side effects in the POMC trial included injection site reaction and hyperpigmentation in all participants, nausea in five participants, and vomiting in three. In the LEPR trial, eleven participants experienced injection site reactions, five had skin disorders, and four reported nausea [54]. A separate clinical trial in obese BBS patients showed that 32.3% achieved weight loss after one year of Setmelanotide treatment. The most common side effect reported by participants was skin hyperpigmentation, occurring in 60.5% of participants [62]. Overall, Semaglutide, Liraglutide, and Setmelanotide represent promising parenteral pharmacological agents that have demonstrated significant clinical efficacy in managing obesity. Ongoing studies and clinical trials continue to evaluate their long-term safety, effectiveness, and potential for broader applications in obesity treatment.

2.4. Tirzepatide

Tirzepatide is the most recent FDA-approved drug for treating type 2 diabetes mellitus (T2DM) and obesity. It is intended for long-term use, with studies assessing its effects over approximately 72 weeks [63,64]. Tirzepatide is a GLP-1 and GIP receptor agonist that binds and activates these receptors, causing decreased appetite and lowering food intake [64]. It was approved by the FDA in November 2023 for use in chronic weight management, specifically in combination with dietary modifications (a reduced calorie diet) and increased physical activity [63]. Tirzepatide’s dual mechanism of action makes it a promising new weight-loss medication option. In a double-blind, randomized, multicenter, placebo-controlled phase 3 trial, patients who received once-weekly Tirzepatide showed significant reductions in body weight [64]. Specifically, patients receiving 10 mg and 15 mg doses of Tirzepatide achieved an average weight loss of 12.8% and 14.7%, respectively, over 72 weeks, demonstrating a therapeutic effect on body weight [64]. In another phase 3 trial, patients with type 2 diabetes were either assigned Tirzepatide or Semaglutide, and those who received Tirzepatide had a superior mean change in glycated hemoglobin [65]. In this trial, patients who received Tirzepatide at doses of either 5 mg, 10 mg, or 15 mg had a mean change in glycated hemoglobin by –2.01, –2.24, or –2.30, respectively, compared to Semaglutide which resulted in a mean difference of –1.86%. Tirzepatide has been studied in patients with only obesity, with obesity and type 2 diabetes, and patients with only diabetes, and in all, it has shown strengths as a practical option for weight loss. An exciting part of Tirzepatide research has been the improvement in insulin responses in patients with type 2 diabetes over Semaglutide [66]. Tirzepatide was shown to improve bodily insulin secretion in patients who had previously been unresponsive to similar medications while causing more considerable weight loss than Semaglutide [66]. In addition to the weight loss findings of these studies, Tirzepatide has shown little increase in adverse events compared to similar medications [67]. Based on the findings of these studies, Tirzepatide is a compelling new drug due to its action on patients’ insulin levels and weight loss results, offering optimism for the future of weight loss medications.
Figure 2 demonstrates the timeline of FDA approvals for weight loss parenteral medications [68,69,70,71,72].

3. Peroral Medications for Weight Loss

In addition to parenteral medications, various peroral medications are also available for weight loss.

3.1. Phentermine

Phentermine is a primary amine, Schedule 4 drug (low risk of abuse or dependence) that was shown in the 1970s to inhibit the metabolism of serotonin by monoamine oxidase but was never labelled as a monoamine oxidase (MAO) inhibitor [73,74]. Phentermine was FDA-approved in 1959, and it became widely used in 1960. It was initially combined with fenfluramine and dexfenfluramine; however, this was discontinued after finding abnormal values amongst 30% of consumers. Later, it was approved in 2012 to be used alone or in combination with topiramate [75].
Pharmacological interventions such as the sympathomimetic amine phentermine (used in some countries longer term under specialist supervision) remain one of the most prescribed short-term treatments for obesity. The drug primarily increases norepinephrine levels within the hypothalamus, leading to decreased appetite and increased satiety signals [76]. Phentermine shows notable effectiveness, with clinical studies demonstrating that 60% of patients lose at least 5% of their total body weight within three months of treatment [77,78,79,80,81]. The medication shows particular promise in the early weeks of treatment, with patients experiencing an average weight loss of 3.6–7.2 kg over 12 weeks [77,78,79,80,81], making it a go-to short-term solution for obesity management. Its regulatory classification as a Schedule 4 drug indicates its controlled but recognized medical use [77,78,79,80,81].
Although its effectiveness has been established, regulatory restrictions respond to safety concerns. Insomnia, dry mouth, and constipation have been reported in 15–30% of subjects as the most frequent adverse effects [79]. However, the cardiovascular effects of increased heart rate and blood pressure are more concerning, prompting a thoughtful selection of patients. These risks on the cardiovascular level have made those patients with uncontrolled hypertension, a known history of hyperthyroidism, or previously diagnosed cardiac disease contraindicated [80].
The similarities between phentermine and amphetamines are such that the Drug Enforcement Administration of America classifies it as a Schedule 4 controlled substance, meaning its prescription use is regulated. There are guidelines concerning treatment that suggest not treating patients for longer than 12 weeks, although some studies demonstrate safety over the long term when used under close medical supervision [81]. For example, patient selection criteria include extensive cardiovascular screening and continuous monitoring of vital signs during treatment. In conclusion, phentermine remains a widely prescribed short-term pharmacological treatment for obesity due to its ability to suppress appetite and induce modest weight loss, particularly in the early weeks of treatment.

3.2. Phentermine/Topiramate (Qsymia)

Topiramate is a hexose derivative 2,3:4,5-di-O-isopropylidene-β-D-fructopyranose, in which the hydroxy group has been converted to the corresponding sulfamate ester. It blocks voltage-dependent sodium channels and is used in treating migraines. Additionally, it has a role as an anticonvulsant and a sodium channel blocker. It was initially approved by the FDA in 1996 and was approved in 2004 for migraine prevention. It was discovered by chance when attempting to formulate an anti-diabetic drug. In 2012, it was approved for combination with phentermine (refer to the previous subtopic for more information) for chronic weight management therapy in adults [82]. Phentermine–topiramate is a Schedule 4 drug [83]. The extended-release fixed-dose combination of phentermine and topiramate represents a significant advancement in obesity pharmacotherapy. This combination combines two pharmacologically complementary mechanisms of action: phentermine, a sympathomimetic amine anorectic, as mentioned in the previous paragraph, works by increasing norepinephrine levels within the hypothalamus, leading to a decrease in appetite; and topiramate, which enhances satiety and energy expenditure [76,84]. This synergistic effect permits the administration of lower dosages needed from each component to avoid adverse effects at decreased therapeutic efficacy.
Clinical trials demonstrate more significant weight loss with the combination than monotherapy with either drug. In the 56-week CONQUER trial of overweight and obese adults (n = 2487), participants receiving the highest approved dose (15/92 mg) experienced mean weight reductions of 9.8% after 56 weeks compared to 1.2% with the placebo [85]. In some subgroup analyses, as much as 70% of patients taking high-dose PHEN/TPM ER achieved ≥5% weight loss, with 48% achieving loss ≥10% from the baseline [86]. The SEQUEL study further established its long-term efficacy, showing sustained weight loss over 2 years and improvements in cardiovascular risk factors [87]. This combination therapy is also associated with beneficial outcomes for obesity-related comorbidities, including notable reductions in sleep apnea severity and significant enhancements in quality-of-life parameters.
Side effects profile revealed dose-dependent adverse events, with the most commonly reported being paresthesia, dry mouth, constipation, and insomnia. Other side effects include cognitive function, affecting attention and memory impairment, necessitating gradual dose titration and regular monitoring [81]. Given its teratogenicity, strict pregnancy testing and contraception requirements must apply to women of childbearing potential. Overall, phentermine–topiramate represents a significant advancement in obesity management, offering enhanced weight loss and metabolic benefits. However, it requires careful patient monitoring to manage its side effects and ensure safe use, particularly in women of reproductive age.

3.3. Bupropion/Naltrexone (Contrave)

Bupropion is an aminoketone propiophenone carrying a tert-butylamino group at position two and a chloro substituent at position three on the phenyl ring. The mechanism of action of bupropion is as a dopamine uptake inhibitor and norepinephrine uptake inhibitor. The physiological effect of bupropion is increased dopamine activity and norepinephrine activity. It is a secondary amino compound, a member of monochlorobenzenes, and an aromatic ketone [88]. The medication was first patented by Burroughs Welcome in 1974 and approved by the FDA in 1985 [89]. Naltrexone is an organic heteropentacyclic compound that is naloxone substituted in which a cyclopropyl methyl group replaces the allyl group attached to the nitrogen. A mu-opioid receptor antagonist, it is used to treat alcohol dependence. It is an organic heteropentacyclic compound, a morphine-like compound, and a member of cyclopropanes. It is a derivative of noroxymorphone that is the N-cyclopropyl methyl congener of naloxone [90]. It was developed in 1963 for alcohol use disorder and opioid dependence, patented in 1967, and received FDA approval in 1984 [91].
The combination of bupropion, a norepinephrine–dopamine reuptake inhibitor, and naltrexone, an opioid receptor antagonist, provides a novel approach to weight management through dual modulation of appetite and reward pathways. Bupropion activates proopiomelanocortin (POMC) neurons of the hypothalamus, suppressing feeding, increasing expenditure and improving glucose tolerance. At the same time, naltrexone blocks β-endorphin-dependent POMC auto-inhibition and thus maintains prolonged activity for anorexigenic pathways, suppressing appetite [92]. At 56 weeks, patients on the prescribed dose in the phase III COR-I study lost an average of 6.1% of their body weight, while those on the placebo lost 1.3% [93]. A post hoc study revealed that the greatest effects occurred within 28 weeks after the start of treatment, with approximately 48% of treated patients seeing weight loss greater than 5% [94]. Patient tolerability varies; 29.8% experienced nausea, while secondary adverse reactions included headache (17.6%) and constipation (19.2%). The high rate of dropouts due to adverse events, approaching 24% in clinical trials, requires cautious patient counselling and slow dose escalation [95]. The combination has selectively found use in patients with mood disorders (in addition to substance abuse or eating disorders) and atypical depression, potentially due to bupropion’s anti-depressant properties. Long-term cardiovascular outcomes have been evaluated through the LIGHT study. However, it was halted ahead of schedule after it fell short of its primary CVD outcome goal to judge long-term cardiovascular safety (for which preliminary data instead suggested no added cardiovascular danger). Blood pressure control remains important because modest increases have been reported, especially in the first 2 weeks of therapy [96]. In conclusion, the combination of bupropion and naltrexone offers an innovative approach to weight management by modulating both appetite and reward pathways by activating POMC neurons and inhibiting opioid receptors. Clinical trials have demonstrated its efficacy, with significant weight loss observed in patients, though tolerability and adherence may be challenged by adverse effects such as nausea, headache, and constipation.

3.4. Orlistat (Alli, Xenical)

Orlistat is a carboxylic ester resulting from the formal condensation of the carboxy group of N-formyl-L-leucine with the hydroxy group of (3S,4S)-3-hexyl-4-[(2S)-2-hydroxytridecyl] oxetane-2-one. It is a beta-lactone, an L-leucine derivative, a member of formamides, and a carboxylic ester. It is a saturated derivative derived from the endogenous lipstatin in Streptomyces toxytricinihas [97]. It has the role of a triacylglycerol inhibitor, bacterial metabolite, fatty acid synthase inhibitor, and anti-obesity agent [98]. The weight loss drug orlistat [Xenical] offers a novel peripheral mechanism in treating obesity as an inhibitor of gastrointestinal lipase. It was initially approved by the US FDA in 1999 as a prescription product [99].
Orlistat has no effects on the central nervous system and reduces dietary fat absorption by up to 30%, which results in a negative energy balance [100,101]. It is now the only FDA-approved weight loss drug for use in teenagers between the ages of 12 and 16. It is available as an over the counter (Alli) and prescription (Xenical) product with strengths of 60 mg and 120 mg, respectively. A 1-year weight loss in the range of 2.9 to 3.4 kg is greater than a placebo after a year of treatment, according to meta-analyses of randomized controlled studies, which show modest but consistent efficacy [101]. The XENDOS study’s four-year randomized data showed that treated individuals had a decreased incidence of new-onset type 2 diabetes and sustained weight loss, with 52.8% losing ≥5% bodyweight in one year [102].
Mechanism of action-related gastrointestinal side effects affect about 15–30% of patients. These include oily spotting, flatus with discharge, and fecal urgency when the fat content in the diet is more than 30% per calorie [103]. As such, patient education regarding dietary modification and fat intake restriction continues to be paramount for both efficacy and tolerability. Long-term safety data is robust, with minimal systemic absorption contributing to a favorable cardiovascular risk profile. Nonetheless, a few cases of significant liver injury have occurred that required monitoring of hepatic function [104]. In conclusion, orlistat provides a unique, non-systemic approach to weight management by inhibiting dietary fat absorption, offering modest but sustained weight loss and a favorable cardiovascular risk profile.

3.5. Metformin (Off-Label Use)

Metformin hydrochloride is a leading biguanide antihyperglycemic agent that manages high blood sugar levels in type 2 diabetes and is derived from Galega officinalis, a plant otherwise known as goat’s rue. Guanidine was first created in the late 1870s and, when administered to rabbits in 1918, was shown to lower blood glucose levels. Following this, guanidine derivatives were proposed as possible treatments for diabetes, including the aminoguanidine glargine and the more powerful diguanide synthalin. Soon after, toxicity was noted, and by the 1930s, they had been overshadowed by insulin, which had been identified in 1922 and was beginning to be manufactured more widely [105]. It was officially approved in 1994 for treating type 2 diabetes and has since become the most widely prescribed medication [106].
Although, most importantly, it is used off-label for managing type 2 diabetes, metformin has shown promising results in different patient populations concerning weight loss. Its mechanisms of action include reduced hepatic glucose production, enhanced peripheral insulin sensitivity, and modulation of the gut microbiome [107]. Clinical studies indicate metformin causes a slight weight loss in at-risk populations (those who are insulin-resistant/overweight). Meta-analyses suggest that, in overweight or obese participants, a reduction of 1.1–2.6 kg occurs compared to placebo over 6–12 months [108]. The most prominent example was the Diabetes Prevention Program, which showed sustained weight loss over 2.8 years and a more significant impact in patients with higher baseline BMIs [109]. Specific populations demonstrating enhanced benefit include individuals with polycystic ovary syndrome (PCOS), prediabetes, and antipsychotic-induced weight gain. The weight loss is also relatively modest, with a mean reduction of 2.7 kg over six months in patients observed for PCOS during metformin therapy, lending balance to any improvements achieved herein and/or previously reported regarding menstrual regularity and metabolic parameters [110].
In addition, gastrointestinal side effects, including diarrhea and nausea, the latter occurring in 20–30% of cases, can be mitigated by gradually increasing dose titration or using extended-release formulations. Its favorable safety profile, long track record in the clinical use, and potential metabolic effects render metformin an intriguing possibility for specific patient populations, especially those with insulin resistance or prediabetic states [111].
In conclusion, while primarily used for type 2 diabetes, metformin’s modest but beneficial effects on weight loss and metabolic health in specific populations—such as those with insulin resistance, PCOS, and prediabetes—make it a promising off-label option, especially given its well-established safety profile and potential for managing obesity-related conditions.

4. Emerging Medications in Clinical Trials

Weight loss medications are an ever-changing field, with several drugs emerging recently. The rise of Semaglutide (Ozempic) has pushed research in this area much further than before. Several promising medications, including Cagrilintide and Bimagrumab, have not yet been fully approved or put on the market, although both have been studied extensively in clinical trials.

4.1. Cagrilintide

Cagrilintide is a long-acting amylin analogue. Amylin is a 37 amino acid peptide and an essential pancreatic hormone for maintaining homeostasis [112,113]. The pancreas releases amylin and insulin, which then travel to the brain and induce a feeling of satiety [114]. Cagrilintide, primarily being evaluated for its role in weight loss treatments, has shown efficacy when combined with other medications, such as Semaglutide [115]. In clinical studies, Cagrilintide was distributed via subcutaneous injection, ensuring consistent therapeutic levels throughout the treatment period. In a systematic review of 76 trials involving weight loss management drugs, combining Cagrilintide and Semaglutide led to the highest reduction in weight, with a mean difference of –14.03 kg during at least 12 weeks [115]. In a randomized, controlled, phase 1b trial, Cagrilintide use reduced mean body weight significantly compared to the placebo group [116]. This trial, based in the US, followed patients over the course of 25 weeks, with 16 weeks of drug escalation, 4 weeks of finalized dose treatment, and 5 weeks of follow up [116]. In this trial, patients who received 1.2 mg and 2.4 mg of Cagrilintide had mean body weight reductions of 15.7% and 17.1%, respectively, compared to the placebo group, with reductions of 9.8% at week 20 [116]. Finally, in a phase 2 clinical trial, the combination of Cagrilintide and Semaglutide showed the most significant reductions in body weight with a mean difference of −15.6% versus Cagrilintide or Semaglutide treatment alone; these yielded −8.1% and −5.1% respectively [112].
Regarding tolerability, Cagrilintide’s adverse effects range from mild to moderate but maintain a low overall risk profile [116]. The adverse events reported surrounding Cagrilintide’s use were mainly gastrointestinal symptoms such as nausea, as well as superficial reactions to the location of the medication administration [117]. Although it is still under investigation, Cagrilintide has shown immense promise and, with more research, may prove to be the next horizon for weight loss management [117].

4.2. Bimagrumab

Bimagrumab is a human monoclonal antibody that binds to the ActRII signaling pathway, blocking it [118]. ActRII signaling blockage via this antibody effectively increases body muscle mass [118]. Bimagrumab is proving to be a novel therapy based on its mechanism of action, which involves impacting not only body fat, but also body muscle mass [119]. Bimagrumab is a class of drugs made via a monoclonal antibody [118]. Monoclonal antibodies are precise drugs, as they are immunoglobulins which are extremely specific for one epitope or antigen [120]. Most weight loss medications on the market target body fat, metabolism, or hormone pathways that can induce or reduce hunger; however, Bimagrumab is original in its mechanism of action by blocking the ActRII signaling as a human anti-ActRII antibody [118]. In clinical settings, Bimagrumab is anticipated to be administered via intravenous infusion, similar to other monoclonal antibody therapies. In a study performed on diet-induced obese mice, Bimagrumab, used in combination with Semaglutide, showed a reduction in fat mass while also showing a ~10% increase in lean mass [121]. In the same study, Semaglutide alone showed significant muscle and fat mass decreases, whereas using Bimagrumab preserved and increased muscle mass [121]. In a phase 2 randomized clinical trial that lasted 48 weeks, Bimigrumab showed a larger decrease in body fat mass than the group who received the placebo [119]. This trial was specifically for adults with type 2 diabetes, BMIs of 28–40, and HbA1c levels of 6.5% to 10.0% [119]. The promising data came from nine different locations in the US and the UK. Due to the protective nature of its mechanism of action, Bimagrumab is generating excitement in the clinical community. It is well tolerated and able to decrease fat mass and uniquely increase muscle mass, thus proving to be a pioneering treatment, especially for those unable to tolerate the mechanism of action of other weight loss medications [122].

5. Medications Withdrawn from the Market

Lorcaserin (Belviq)

Lorcaserin is a medication that was used for weight loss until it was pulled from the market after the findings of a clinical trial which showed increased development of cancers in patients taking the medication. Lorcaserin works as a serotonergic agonist, specific to the 5HT2C receptor [123]. The 5HT2C receptor is a serotonin receptor that controls crucial functions in maintaining homeostasis, such as food intake [124]. Although initial research showed promise, Lorcaserin is no longer on the market as of 2020 [125]. In 2012, Lorcaserin was approved for patients with obesity as a medication to use while also undergoing lifestyle modifications, such as more physical activity along with a calorie-deficient diet [125]. As a part of the approval process, the FDA required the manufacturer of Lorcaserin to conduct a randomized, double-blind, placebo-controlled clinical trial to continue monitoring the drug’s safety [126]. This trial showed that 7.7% of the Lorcaserin group developed cancers as opposed to 7.1% in the placebo group [126]. Lorcaserin contributed to an overall increase in cancers, specifically lung, pancreatic, and colorectal cancers [126]. Although a 0.6% increase in cancers may not sound significant, 462 patients treated with Lorcaserin developed 520 primary malignancies compared to the placebo group, which had 423 patients who developed 470 primary cancers [126]. Lorcaserin was advised to be pulled from the market by the FDA in 2020 due to the findings of this clinical trial, and it has not been available since [126]. It is paramount to the safety of patients that we continue to monitor drugs, even after they have been brought to market.

6. Artificial Intelligence (AI) in Weight Loss Treatment

AI has become a novel part of everyday life, from ChatGPT to Snapchat’s AI for users. AI is revolutionizing the scientific world, especially within the medical field. AI is currently under investigation for implementation in diagnostic techniques, data gathering, and personalized medicine, which may be essential in weight loss management [127]. As each patient may require a different and precise approach, AI could be essential in precision medicine for weight loss. However, targeted approaches to weight loss management with AI would need to be examined clinically before being released to the public. Currently, the main categories of weight loss interventions are lifestyle modifications, including changing diet, physical activity, mental/emotional well-being, medication, and surgery. AI has enormous potential to help patients tailor their lifestyle changes to their needs. A scoping review on AI use in weight loss found that AI can assist in personalized weight loss when patients input the correct metrics (goals, lifestyle behaviors, etc.) [128]. However, inputting all the proper metrics brings up a downside to using AI: taking the time to input all the data is the patient’s responsibility. Most of the research performed on AI use in weight loss management has said that although AI has vast potential in this area, it has yet to be implemented in such a way as to make a significant impact [129]. In a promising study conducted in 2022, an AI robot could help the user make weight loss decisions [130]. In another study performed in 2023, the results showed that weight-management experts agreed with the modifications and predictions made by the machine learning model, which would prove an exciting leap for AI use in weight loss management programs [131].

7. Comparison Between Different Weight Loss Management Modalities

Table 1 provides a comprehensive comparison of different weight management modalities, including lifestyle interventions, pharmacotherapy, bariatric surgery, and endoscopic procedures. It highlights their mechanisms, efficacy, suitability, and limitations to assist clinicians and researchers in making informed decisions.

8. Safety, Efficacy, and Regulatory Considerations

Generally, the current anti-obesity medications (AOMs) have shown a significant effect on weight loss, as one year of AOMs can result in at least 5% weight reduction, which can improve many obesity-associated comorbidities [171]. Also, they achieved the same weight reduction percentage in 44–75% of 29,018 patients [171]. Nevertheless, the most commonly reported adverse effects of AOMs are dry mouth, nausea, diarrhea, and constipation, which are generally manageable [172,173,174,175,176]. On the other hand, some serious adverse events have been associated with these drugs, such as cardiovascular disease, acute kidney failure, and kidney injury [177,178]. There are some contraindications for the use of AOMs, mainly for pregnant or breastfeeding women and patients with severe renal or hepatic impairments. On the other hand, AOMs can be prescribed with extra caution in patients with non-severe renal or hepatic impairments and elderly patients. However, there are limited studies on the latter [19]. Since most clinical trials monitor drug-induced risks for only one year, we recommend further investigations of drug safety in the long term.
Regulatory oversight of AOMs is crucial to ensure their safety and efficacy before and after approval, and it involves multiple steps, from clinical trials to drug approval and post-market surveillance. According to the Food and Drug Administration (FDA), the first two phases of clinical trials should evaluate the pharmacokinetics of AOMs, their maximum doses at which they become active compared to placebo, and their ability to achieve weight loss ≥ 5% of the baseline weight. Then, phase 3 clinical trials examine the efficiency and safety profile of AOMs in randomized, double-blinded, and placebo-controlled trials. The effect on obesity-related morbidities should also be assessed throughout all phases [179]. The efficacy criteria for AOMs are either that the difference in average weight loss between the drug- and placebo-treated groups being ≥5% or the drug-treated group achieving a weight loss of ≥5% from baseline, with at least 35% of participants reaching this threshold, double the proportion of the placebo-treated group [179,180]. Both outcomes should follow one year of AOMs and be statistically significant [179,180]. The safety estimation should be based on 3000 participants in a randomized one-year trial, with at least 1500 subjects given a placebo [179]. Furthermore, given the critical role of post-market surveillance, we advise clinicians to report adverse drug reactions (ADRs) to their local regulatory agency and to encourage patients to inform their healthcare providers of any suspect ADRs.
Some recommendations can be followed to improve access to AOMs. First, they should be covered by health insurance as a core element rather than a non-essential extra. In addition, the overpricing of AOMs can hinder their access to patients and impose financial burdens on them and the healthcare system. Therefore, AOMs pricing should reflect their effectiveness and the uncertainty surrounding long-term benefits, with initial prices set reasonably low until further evidence validates their long-term benefits. Furthermore, since not all people are qualified for AOMs, it is crucial to establish a set of criteria that informs treatment allocation for those who are indicated. Although BMI remains the main measure for assessing obesity, experts emphasize the need for additional measures of obesity severity for routine clinical use to aid in identifying individuals at an increased risk of obesity-related complications, allowing for better prioritization of treatment and subsequently expanding access to AOMs among those who need them most [181].
Moreover, medical specialty societies are recommended to create and distribute educational materials that allow AOMs prescription to qualified patients by a wide range of clinicians rather than weight loss specialists exclusively [181]. When it comes to prescribing AOMs, the individual response to AOMs is variable, so prescribers should account for the MOA, safety, adverse effects, contraindications, drug interactions, and route of administration. Also, affordability should be considered with patients when selecting from available options and re-evaluated periodically. If AOMs fail to achieve clinically meaningful weight loss, other factors like the dose, adherence, behavioral change challenges, and mental or medical conditions should be taken into account, and the prescribed AOM should be stopped if it does not result in ≥5% weight loss within 3 months (4 months for naltrexone/bupropion combination therapy). Currently, there is no available method to predict which AOM will be the most effective for an individual patient. However, advancements in the field of personalized medicine, such as hormonal and genetic profiling, could make this possible. Thus, investment in these areas would be highly valuable [182].

9. Conclusions

With the rising prevalence of obesity and its associated comorbidities, there is a growing motivation to develop innovative solutions to address this issue. One of them is lifestyle modification, which is not always sufficient to resolve obesity, necessitating more potent interventions like anti-obesity medications AOMs and surgical procedures.
Developing new AOMs has been challenged by the efficacy and safety profiles that AOMs must achieve to be considered safe and effective. Over the past 20 years, many AOMs have emerged with mechanisms of actions that reduce hunger, promote satiety, and increase energy expenditure, leading to weight loss. Some of these drugs were made available in parenteral and peroral forms, and others were approved as combination drugs. However, some were withdrawn from the market, notably Lorcaserin, due to the risk of cancer. AOMs have managed to achieve a significant reduction in obesity and its associated complications, including diabetes, cardiovascular diseases, and sleep apnea. However, there are some side effects associated with these drugs, commonly reported as dry mouth, insomnia, and gastrointestinal symptoms, or severe adverse effects like pancreatitis, gallbladder disease, or liver injury. Thus, regulatory oversight is critical to ensure high standards of safety and efficacy.
Moreover, bariatric surgeries and endoscopic weight loss procedures have become viable options for patients requiring interventions beyond AOMs and lifestyle changes. The landscape of obesity management is rapidly evolving, with novel AOMs in clinical trials showing promising safety and efficacy profiles. Additionally, the emerging artificial intelligence (AI) has demonstrated potential benefits by personalizing weight loss interventions for patients and monitoring their progression.

Author Contributions

Conceptualization, H.A.L.; Writing—original draft preparation, A.A.L., A.A., R.R., M.-L.S., H.A., S.D. and H.A.L.; Figures, S.D.; Supervision, H.A.L. All authors have read and agreed to the published version of the manuscript.

Funding

The authors would like to thank the Oman College of Health Sciences (OCHS) for covering the article processing charges (APC) for this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
5HT2C5-hydroxytryptamine 2C receptor
AACEAmerican Association of Clinical Endocrinologists
ACCAmerican College of Cardiology
ACEAmerican College of Endocrinology
ADRsAdverse Drug Reactions
AHAAmerican Heart Association
AIArtificial Intelligence
AOMsAnti-Obesity Medications
AgRPAgouti-Related Peptide
BBSBardet–Biedl Syndrome
BMIBody Mass Index
CARTCocaine- and Amphetamine-Regulated Transcript
CVDsCardiovascular Diseases
DALYsDisability-Adjusted Life Years
DBPDiastolic Blood Pressure
DPP-4Dipeptidyl Peptidase-4
FDAU.S. Food and Drug Administration
GLP-1Glucagon-Like Peptide-1
HDLHigh-Density Lipoprotein
LEPRLeptin Receptor
MAOMonoamine Oxidase
MC4RMelanocortin-4 Receptor
NPYNeuropeptide Y
PCOSPolycystic Ovary Syndrome
PCSK1Proprotein Convertase Subtilisin/Kexin Type 1
POMCProopiomelanocortin
QALYQuality-Adjusted Life Year
RCTsRandomized Control Trials
SBPSystolic Blood Pressure
T2DType 2 Diabetes
TBWTotal Body Weight
TOSThe Obesity Society
TRGTriglycerides
WHOWorld Health Organization

References

  1. Lin, X.; Li, H. Obesity: Epidemiology, pathophysiology, and therapeutics. Front. Endocrinol. 2021, 12, 706978. [Google Scholar] [CrossRef] [PubMed]
  2. World Health Organization. A Healthy Lifestyle—WHO Recommendations. Available online: https://www.who.int/europe/news-room/fact-sheets/item/a-healthy-lifestyle---who-recommendations (accessed on 27 February 2025).
  3. NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: A pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults. Lancet 2017, 390, 2627–2642. [Google Scholar] [CrossRef] [PubMed]
  4. Kelly, T.; Yang, W.; Chen, C.S.; Reynolds, K.; He, J. Global burden of obesity in 2005 and projections to 2030. Int. J. Obes. 2008, 32, 1431–1437. [Google Scholar] [CrossRef] [PubMed]
  5. Chooi, Y.C.; Ding, C.; Magkos, F. The epidemiology of obesity. Metabolism 2019, 92, 6–10. [Google Scholar] [CrossRef]
  6. Ochoa-Moreno, I.; Taheem, R.; Woods-Townsend, K.; Chase, D.; Godfrey, K.M.; Modi, N.; Hanson, M. Projected health and economic effects of the increase in childhood obesity during the COVID-19 pandemic in England: The potential cost of inaction. PLoS ONE 2024, 19, e0296013. [Google Scholar] [CrossRef]
  7. World Health Organization. Obesity and Overweight. Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 27 February 2025).
  8. Dai, H.; Alsalhe, T.A.; Chalghaf, N.; Riccò, M.; Bragazzi, N.L.; Wu, J. The global burden of disease attributable to high body mass index in 195 countries and territories, 1990–2017: An analysis of the Global Burden of Disease Study. PLoS Med. 2020, 17, e1003198. [Google Scholar] [CrossRef] [PubMed]
  9. Chen, Y.; Ma, L.; Han, Z.; Xiong, P. The global burden of disease attributable to high body mass index in 204 countries and territories: Findings from 1990 to 2019 and predictions to 2035. Diabetes Obes. Metab. 2024, 26, 3998–4010. [Google Scholar] [CrossRef] [PubMed]
  10. Aras, M.; Tchang, B.G.; Pape, J. Obesity and diabetes. Nurs. Clin. N. Am. 2021, 56, 527–541. [Google Scholar] [CrossRef]
  11. Rippe, J.M.; Crossley, S.; Ringer, R. Obesity as a chronic disease: Modern medical and lifestyle management. J. Am. Diet. Assoc. 1998, 98, S9–S15. [Google Scholar] [CrossRef]
  12. Field, A.E.; Coakley, E.H.; Must, A.; Spadano, J.L.; Laird, N.; Dietz, W.H.; Rimm, E.; Colditz, G.A. Impact of overweight on the risk of developing common chronic diseases during a 10-year period. Arch. Intern. Med. 2001, 161, 1581–1586. [Google Scholar] [CrossRef]
  13. Wang, T.J.; Parise, H.; Levy, D.; D’Agostino, R.B., Sr.; Wolf, P.A.; Vasan, R.S.; Benjamin, E.J. Obesity and the risk of new-onset atrial fibrillation. JAMA 2004, 292, 2471–2477. [Google Scholar] [CrossRef]
  14. Bogers, R.P.; Bemelmans, W.J.; Hoogenveen, R.T.; Boshuizen, H.C.; Woodward, M.; Knekt, P.; van Dam, R.M.; Hu, F.B.; Visscher, T.L.; Menotti, A.; et al. Association of overweight with increased risk of coronary heart disease partly independent of blood pressure and cholesterol levels: A meta-analysis of 21 cohort studies including more than 300,000 persons. Arch. Intern. Med. 2007, 167, 1720–1728. [Google Scholar] [CrossRef] [PubMed]
  15. Poirier, P.; Giles, T.D.; Bray, G.A.; Hong, Y.; Stern, J.S.; Pi-Sunyer, F.X.; Eckel, R.H.; Franklin, B.A.; Hunter, G.; Milani, R.V.; et al. Obesity and cardiovascular disease: Pathophysiology, evaluation, and effect of weight loss. Circulation 2006, 113, 898–918. [Google Scholar] [CrossRef] [PubMed]
  16. Magkos, F.; Fraterrigo, G.; Yoshino, J.; Luecking, C.; Kirbach, K.; Kelly, S.C.; de las Fuentes, L.; He, S.; Okunade, A.L.; Patterson, B.W.; et al. Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity. Cell Metab. 2016, 23, 591–601. [Google Scholar] [CrossRef] [PubMed]
  17. Bays, H.E.; Toth, P.P.; Kris-Etherton, P.M.; Abate, N.; Aronne, L.J.; Brown, W.V.; Gonzalez-Campoy, J.M.; Jones, S.R.; Kumar, R.; La Forge, R.; et al. Obesity, adiposity, and dyslipidemia: A consensus statement from the National Lipid Association. J. Clin. Lipidol. 2013, 7, 304–383. [Google Scholar] [CrossRef]
  18. Wing, R.R.; Lang, W.; Wadden, T.A.; Safford, M.; Knowler, W.C.; Bertoni, A.G.; Hill, J.O.; Brancati, F.L.; Peters, A.; Wagenknecht, L.; et al. Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes Care 2011, 34, 1481–1486. [Google Scholar] [CrossRef] [PubMed]
  19. Garvey, W.T.; Mechanick, J.I.; Brett, E.M.; Garber, A.J.; Hurley, D.L.; Jastreboff, A.M.; Nadolsky, K.; Pessah-Pollack, R.; Plodkowski, R.; Bays, H.E.; et al. Comprehensive clinical practice guidelines for medical care of patients with obesity. Endocr. Pract. 2016, 22, 842–884. [Google Scholar] [CrossRef] [PubMed]
  20. Jensen, M.D.; Ryan, D.H.; Apovian, C.M.; Ard, J.D.; Comuzzie, A.G.; Donato, K.A.; Hu, F.B.; Hubbard, V.S.; Jakicic, J.M.; Kushner, R.F.; et al. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults. J. Am. Coll. Cardiol. 2014, 63, 2985–3023. [Google Scholar] [CrossRef]
  21. Wharton, S.; Lau, D.C.W.; Vallis, M.; Sharma, A.M.; Biertho, L.; Campbell-Scherer, D.; Adamo, K.; Alberga, A.; Bell, R.; Boulé, N.; et al. Obesity in adults: A clinical practice guideline. CMAJ 2020, 192, E875–E891. [Google Scholar] [CrossRef]
  22. Garber, A.J. Long-acting glucagon-like peptide 1 receptor agonists: A review of their efficacy and tolerability. Diabetes Care 2011, 34, S279–S284. [Google Scholar] [CrossRef]
  23. Scheen, A.J. Le médicament du mois. Le liraglutide à la dose de 3 mg (Saxenda): Indication dans le traitement de l’obésité. Rev. Med. Liege 2016, 71, 256–261. [Google Scholar] [PubMed]
  24. Knudsen, L.B.; Nielsen, P.F.; Huusfeldt, P.O.; Johansen, N.L.; Madsen, K.; Pedersen, F.Z.; Thøgersen, H.; Wilken, M.; Agersø, H. Potent derivatives of glucagon-like peptide-1 with pharmacokinetic properties suitable for once daily administration. J. Med. Chem. 2000, 43, 1664–1669. [Google Scholar] [CrossRef] [PubMed]
  25. Kim, W.; Egan, J.M. The role of incretins in glucose homeostasis and diabetes treatment. Pharmacol. Rev. 2008, 60, 470–512. [Google Scholar] [CrossRef]
  26. Garvey, W.T.; Birkenfeld, A.L.; Dicker, D.; Mingrone, G.; Pedersen, S.D.; Satylganova, A.; Skovgaard, D.; Sugimoto, D.; Jensen, C.; Mosenzon, O. Efficacy and safety of liraglutide 3.0 mg in individuals with overweight or obesity and type 2 diabetes treated with basal insulin: The SCALE Insulin randomized controlled trial. Diabetes Care 2020, 43, 1085–1093. [Google Scholar] [CrossRef] [PubMed]
  27. Tamayo-Trujillo, R.; Ruiz-Pozo, V.A.; Cadena-Ullauri, S.; Guevara-Ramírez, P.; Paz-Cruz, E.; Zambrano-Villacres, R.; Simancas-Racines, D.; Zambrano, A.K. Molecular mechanisms of semaglutide and liraglutide as a therapeutic option for obesity. Front. Nutr. 2024, 11, 1398059. [Google Scholar] [CrossRef] [PubMed]
  28. Inoue, K.; Maeda, N.; Fujishima, Y.; Fukuda, S.; Nagao, H.; Yamaoka, M.; Hirata, A.; Nishizawa, H.; Funahashi, T.; Shimomura, I. Long-term impact of liraglutide, a glucagon-like peptide-1 (GLP-1) analogue, on body weight and glycemic control in Japanese type 2 diabetes: An observational study. Diabetol. Metab. Syndr. 2014, 6, 95. [Google Scholar] [CrossRef] [PubMed]
  29. Imbernon, M.; Saponaro, C.; Helms, H.C.C.; Sanchez-Aguilera, P.; García-Cáceres, C.; Secher, A.; Ferrés-Coy, A.; Díaz-Delfín, J.; Hebert-Chatelain, E.; Tena-Sempere, M.; et al. Tanycytes control hypothalamic liraglutide uptake and its anti-obesity actions. Cell Metab. 2022, 34, 1054–1063.e7. [Google Scholar] [CrossRef]
  30. Secher, A.; Jelsing, J.; Baquero, A.F.; Hecksher-Sørensen, J.; Cowley, M.A.; Dalbøge, L.S.; Hansen, G.; Grove, K.L.; Pyke, C.; Raun, K.; et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J. Clin. Invest. 2014, 124, 4473–4488. [Google Scholar] [CrossRef]
  31. Pi-Sunyer, X.; Astrup, A.; Fujioka, K.; Greenway, F.; Halpern, A.; Krempf, M.; Lau, D.C.; le Roux, C.W.; Violante Ortiz, R.; Jensen, C.B.; et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N. Engl. J. Med. 2015, 373, 11–22. [Google Scholar] [CrossRef]
  32. Blackman, A.; Foster, G.D.; Zammit, G.; Rosenberg, R.; Aronne, L.; Wadden, T.; Claudius, B.; Jensen, C.B.; Mignot, E. Effect of liraglutide 3.0 mg in individuals with obesity and moderate or severe obstructive sleep apnea: The SCALE Sleep Apnea randomized clinical trial. Int. J. Obes. 2016, 40, 1310–1319. [Google Scholar] [CrossRef]
  33. Elkind-Hirsch, K.E.; Chappell, N.; Shaler, D.; Storment, J.; Bellanger, D.L. Liraglutide 3 mg on weight, body composition, and hormonal and metabolic parameters in women with obesity and polycystic ovary syndrome: A randomized placebo-controlled phase 3 study. Fertil. Steril. 2022, 118, 371–381. [Google Scholar] [CrossRef] [PubMed]
  34. Wadden, T.A.; Tronieri, J.S.; Sugimoto, D.; Lund, M.T.; Auerbach, P.; Jensen, C.; Rubino, D. Liraglutide 3.0 mg and intensive behavioral therapy (IBT) for obesity in primary care: The SCALE IBT randomized controlled trial. Obesity 2020, 28, 529–536. [Google Scholar] [CrossRef]
  35. Astrup, A.; Rössner, S.; Van Gaal, L.; Rissanen, A.; Niskanen, L.; Al Hakim, M.; Madsen, J.; Rasmussen, M.F.; Lean, M.E.; Wilding, J.P.H.; et al. Effects of liraglutide in the treatment of obesity: A randomised, double-blind, placebo-controlled study. Lancet 2009, 374, 1606–1616. [Google Scholar] [CrossRef]
  36. Astrup, A.; Carraro, R.; Finer, N.; Harper, A.; Kunesova, M.; Lean, M.E.; Niskanen, L.; Rasmussen, M.F.; Rissanen, A.; Rössner, S.; et al. Safety, tolerability, and sustained weight loss over 2 years with the once-daily human GLP-1 analog, liraglutide. Int. J. Obes. 2012, 36, 843–854. [Google Scholar] [CrossRef] [PubMed]
  37. Rubino, D.M.; Greenway, F.L.; Khalid, U.; O’Neil, P.M.; Rosenstock, J.; Sørrig, R.; Wadden, T.A.; Wizert, A.; Garvey, W.T.; Davies, M.; et al. Effect of weekly subcutaneous semaglutide vs daily liraglutide on body weight in adults with overweight or obesity without diabetes: The STEP 8 randomized clinical trial. JAMA 2022, 327, 138–150. [Google Scholar] [CrossRef] [PubMed]
  38. Aldhaleei, W.A.; Abegaz, T.M.; Bhagavathula, A.S. Glucagon-like peptide-1 receptor agonists associated gastrointestinal adverse events: A cross-sectional analysis of the National Institutes of Health All of Us cohort. Pharmaceuticals 2024, 17, 199. [Google Scholar] [CrossRef] [PubMed]
  39. Franks, A.S.; Lee, P.H.; George, C.M. Pancreatitis: A potential complication of liraglutide? Ann. Pharmacother. 2012, 46, 1547–1553. [Google Scholar] [CrossRef]
  40. Chao, A.M.; Tronieri, J.S.; Amaro, A.; Wadden, T.A. Semaglutide for the treatment of obesity. Trends Cardiovasc. Med. 2023, 33, 159–166. [Google Scholar] [CrossRef]
  41. Singh, G.; Krauthamer, M.; Bjalme-Evans, M. Wegovy (semaglutide): A new weight loss drug for chronic weight management. J. Investig. Med. 2022, 70, 5–13. [Google Scholar] [CrossRef]
  42. Amaro, A.; Sugimoto, D.; Wharton, S. Efficacy and safety of semaglutide for weight management: Evidence from the STEP program. Postgrad. Med. 2022, 134 (Suppl. S1), 5–17. [Google Scholar] [CrossRef]
  43. Blundell, J.; Finlayson, G.; Axelsen, M.; Flint, A.; Gibbons, C.; Kvist, T.; Hjerpsted, J.B. Effects of once-weekly semaglutide on appetite, energy intake, control of eating, food preference, and body weight in subjects with obesity. Diabetes Obes. Metab. 2017, 19, 1242–1251. [Google Scholar] [CrossRef] [PubMed]
  44. Drucker, D.J. The GLP-1 journey: From discovery science to therapeutic impact. J. Clin. Investig. 2024, 134, e175634. [Google Scholar] [CrossRef] [PubMed]
  45. Friedrichsen, M.; Breitschaft, A.; Tadayon, S.; Wizert, A.; Skovgaard, D. The effect of semaglutide 2.4 mg once weekly on energy intake, appetite, control of eating, and gastric emptying in adults with obesity. Diabetes Obes. Metab. 2021, 23, 754–762. [Google Scholar] [CrossRef] [PubMed]
  46. Smith, N.K.; Hackett, T.A.; Galli, A.; Flynn, C.R. GLP-1: Molecular mechanisms and outcomes of a complex signaling system. Neurochem. Int. 2019, 128, 94–105. [Google Scholar] [CrossRef]
  47. Sorli, C.; Harashima, S.I.; Tsoukas, G.M.; Unger, J.; Karsbøl, J.D.; Hansen, T.; Bain, S.C. Efficacy and safety of once-weekly semaglutide monotherapy versus placebo in patients with type 2 diabetes (SUSTAIN 1): A double-blind, randomised, placebo-controlled, parallel-group, multinational, multicentre phase 3a trial. Lancet Diabetes Endocrinol. 2017, 5, 251–260. [Google Scholar] [CrossRef] [PubMed]
  48. Rodbard, H.W.; Lingvay, I.; Reed, J.; de la Rosa, R.; Rose, L.; Sugimoto, D.; Araki, E.; Chu, P.L.; Wijayasinghe, N.; Norwood, P. Semaglutide added to basal insulin in type 2 diabetes (SUSTAIN 5): A randomized, controlled trial. J. Clin. Endocrinol. Metab. 2018, 103, 2291–2301. [Google Scholar] [CrossRef]
  49. Zinman, B.; Bhosekar, V.; Busch, R.; Holst, I.; Ludvik, B.; Thielke, D.; Thrasher, J.; Woo, V.; Philis-Tsimikas, A. Semaglutide once weekly as add-on to SGLT-2 inhibitor therapy in type 2 diabetes (SUSTAIN 9): A randomised, placebo-controlled trial. Lancet Diabetes Endocrinol. 2019, 7, 356–367. [Google Scholar] [CrossRef]
  50. Pratley, R.; Amod, A.; Hoff, S.T.; Kadowaki, T.; Lingvay, I.; Nauck, M.; Pedersen, K.B.; Saugstrup, T.; Meier, J.J.; Bain, S.C.; et al. Oral semaglutide versus subcutaneous liraglutide and placebo in type 2 diabetes (PIONEER 4): A randomised, double-blind, phase 3a trial. Lancet 2019, 394, 39–50. [Google Scholar] [CrossRef]
  51. Capehorn, M.S.; Catarig, A.M.; Furberg, J.K.; Janez, A.; Price, H.C.; Tadayon, S.; Vergès, B.; Marre, M. Efficacy and safety of once-weekly semaglutide 1.0 mg vs once-daily liraglutide 1.2 mg as add-on to 1–3 oral antidiabetic drugs in subjects with type 2 diabetes (SUSTAIN 10). Diabetes Metab. 2020, 46, 100–109. [Google Scholar] [CrossRef]
  52. Weghuber, D.; Barrett, T.; Barrientos-Pérez, M.; Gies, I.; Hesse, D.; Jeppesen, O.K.; Kelly, A.S.; Mastrandrea, L.D.; Sørrig, R.; Arslanian, S.; et al. Once-weekly semaglutide in adolescents with obesity. N. Engl. J. Med. 2022, 387, 2245–2257. [Google Scholar] [CrossRef]
  53. Nagendra, L.; Bg, H.; Sharma, M.; Dutta, D. Semaglutide and cancer: A systematic review and meta-analysis. Diabetes Metab. Syndr. 2023, 17, 102834. [Google Scholar] [CrossRef] [PubMed]
  54. Clément, K.; van den Akker, E.; Argente, J.; Bahm, A.; Chung, W.K.; Connors, H.; De Waele, K.; Farooqi, I.S.; Gonneau-Lejeune, J.; Gordon, G.; et al. Efficacy and safety of setmelanotide, an MC4R agonist, in individuals with severe obesity due to LEPR or POMC deficiency: Single-arm, open-label, multicentre, phase 3 trials. Lancet Diabetes Endocrinol. 2020, 8, 960–970. [Google Scholar] [CrossRef] [PubMed]
  55. Pressley, H.; Cornelio, C.K.; Adams, E.N. Setmelanotide: A Novel Targeted Treatment for Monogenic Obesity. J. Pharm. Technol. 2022, 38, 368–373. [Google Scholar] [CrossRef] [PubMed]
  56. Ayers, K.L.; Glicksberg, B.S.; Garfield, A.S.; Longerich, S.; White, J.A.; Yang, P.; Du, L.; Chittenden, T.W.; Gulcher, J.R.; Roy, S.; et al. Melanocortin 4 receptor pathway dysfunction in obesity: Patient stratification aimed at MC4R agonist treatment. J. Clin. Endocrinol. Metab. 2018, 103, 2601–2612. [Google Scholar] [CrossRef] [PubMed]
  57. Markham, A. Setmelanotide: First approval. Drugs 2021, 81, 397–403. [Google Scholar] [CrossRef]
  58. Han, J.C.; Rasmussen, M.C.; Forte, A.R.; Schrage, S.B.; Zafar, S.K.; Haqq, A.M. Management of monogenic and syndromic obesity. Gastroenterol. Clin. N. Am. 2023, 52, 733–750. [Google Scholar] [CrossRef]
  59. Hammad, M.M.; Mohammad, A.; Alam-Eldin, N.; Madhu, D.; Al-Mulla, F.; Abu-Farha, M.; Abubaker, J. Structural analysis of setmelanotide binding to MC4R variants in comparison to wild-type receptor. Life Sci. 2022, 307, 120857. [Google Scholar] [CrossRef]
  60. Yeo, G.S.H.; Chao, D.H.M.; Siegert, A.M.; Koerperich, Z.M.; Ericson, M.D.; Simonds, S.E.; Larson, C.M.; Luquet, S.; Clarke, I.; Sharma, S.; et al. The melanocortin pathway and energy homeostasis: From discovery to obesity therapy. Mol. Metab. 2021, 48, 101206. [Google Scholar] [CrossRef]
  61. Chakhtoura, M.; Haber, R.; Ghezzawi, M.; Rhayem, C.; Tcheroyan, R.; Mantzoros, C.S. Pharmacotherapy of obesity: An update on the available medications and drugs under investigation. EClinicalMedicine 2023, 58, 101882. [Google Scholar] [CrossRef]
  62. Haqq, A.M.; Chung, W.K.; Dollfus, H.; Haws, R.M.; Martos-Moreno, G.Á.; Poitou, C.; Yanovski, J.A.; Mittleman, R.S.; Yuan, G.; Forsythe, E.; et al. Efficacy and safety of setmelanotide, a melanocortin-4 receptor agonist, in patients with Bardet-Biedl syndrome and Alström syndrome: A multicentre, randomised, double-blind, placebo-controlled, phase 3 trial with an open-label period. Lancet Diabetes Endocrinol. 2022, 10, 859–868. [Google Scholar] [CrossRef]
  63. FDA Approves New Medication for Chronic Weight Management. FDA Press Release. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-new-medication-chronic-weight-management (accessed on 27 February 2025).
  64. Garvey, W.T.; Frias, J.P.; Jastreboff, A.M.; le Roux, C.W.; Sattar, N.; Aizenberg, D.; Mao, H.; Zhang, S.; Ahmad, N.N.; Bunck, M.C.; et al. Tirzepatide once weekly for the treatment of obesity in people with type 2 diabetes (SURMOUNT-2): A double-blind, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 2023, 402, 613–626. [Google Scholar] [CrossRef]
  65. Frías, J.P.; Davies, M.J.; Rosenstock, J.; Pérez Manghi, F.C.; Fernández Landó, L.; Bergman, B.K.; Liu, B.; Cui, X.; Brown, K.; SURPASS-2 Investigators; et al. Tirzepatide versus semaglutide once weekly in patients with type 2 diabetes. N. Engl. J. Med. 2021, 385, 503–515. [Google Scholar] [CrossRef]
  66. Nauck, M.A.; D’Alessio, D.A. Tirzepatide, a dual GIP/GLP-1 receptor co-agonist for the treatment of type 2 diabetes with unmatched effectiveness regarding glycaemic control and body weight reduction. Cardiovasc. Diabetol. 2022, 21, 169. [Google Scholar] [CrossRef] [PubMed]
  67. Tan, B.; Pan, X.H.; Chew, H.S.J.; Goh, R.S.J.; Lin, C.; Anand, V.V.; Lee, E.C.Z.; Chan, K.E.; Kong, G.; Ong, C.E.Y.; et al. Efficacy and safety of tirzepatide for treatment of overweight or obesity: A systematic review and meta-analysis. Int. J. Obes. 2023, 47, 677–685. [Google Scholar] [CrossRef]
  68. Nauck, M.A.; Quast, D.R.; Wefers, J.; Meier, J.J. GLP-1 receptor agonists in the treatment of type 2 diabetes—State-of-the-art. Mol. Metab. 2021, 46, 101102. [Google Scholar] [CrossRef] [PubMed]
  69. Myerson, M.; Paparodis, R.D. Pharmacotherapy of weight-loss and obesity with a focus on GLP-1 receptor agonists. J. Clin. Pharmacol. 2024, 64, 1204–1221. [Google Scholar] [CrossRef] [PubMed]
  70. Abdi Beshir, S.; Ahmed Elnour, A.; Soorya, A.; Parveen Mohamed, A.; Sir Loon Goh, S.; Hussain, N.; Al Haddad, A.H.I.; Hussain, F.; Yousif Khidir, I.; Abdelnassir, Z. A narrative review of approved and emerging anti-obesity medications. Saudi Pharm. J. 2023, 31, 101757. [Google Scholar] [CrossRef]
  71. Idrees, Z.; Cancarevic, I.; Huang, L. FDA-Approved Pharmacotherapy for Weight Loss Over the Last Decade. Cureus 2022, 14, e29262. [Google Scholar] [CrossRef]
  72. Pan, X.H.; Tan, B.; Chin, Y.H.; Lee, E.C.Z.; Kong, G.; Chong, B.; Kueh, M.; Khoo, C.M.; Mehta, A.; Majety, P.; et al. Efficacy and safety of tirzepatide, GLP-1 receptor agonists, and other weight loss drugs in overweight and obesity: A network meta-analysis. Obesity 2024, 32, 840–856. [Google Scholar] [CrossRef]
  73. Mayo Clinic. Phentermine Weight-Loss Drug: Know the Side Effects. Available online: https://www.mayoclinic.org/healthy-lifestyle/weight-loss/expert-answers/phentermine/faq-20057940 (accessed on 27 February 2025).
  74. Ulus, I.H.; Maher, T.J.; Wurtman, R.J. Characterization of phentermine and related compounds as monoamine oxidase (MAO) inhibitors. Biochem. Pharmacol. 2000, 59, 1611–1621. [Google Scholar] [CrossRef]
  75. DrugBank. Phentermine. Available online: https://go.drugbank.com/drugs/DB00191 (accessed on 27 February 2025).
  76. Hendricks, E.J.; Rothman, R.B.; Greenway, F.L. How physician obesity specialists use drugs to treat obesity. Obesity 2019, 27, 725–733. [Google Scholar] [CrossRef]
  77. Vivus. Qsymia (Phentermine and Topiramate Extended-Release) Capsules, Prescribing Information; Vivus, Inc.: Mountain View, CA, USA, 2012. [Google Scholar]
  78. Munro, J.F.; MacCuish, A.C.; Wilson, E.M.; Duncan, L.J. Comparison of continuous and intermittent anorectic therapy in obesity. BMJ 2018, 1, 352–354. [Google Scholar] [CrossRef] [PubMed]
  79. Apovian, C.M.; Aronne, L.J.; Bessesen, D.H.; McDonnell, M.E.; Murad, M.H.; Pagotto, U.; Ryan, D.H.; Still, C.D.; Endocrine Society Clinical Guidelines Subcommittee; Endocrine Society Obesity Task Force; et al. Pharmacological management of obesity: An Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2015, 100, 342–362. [Google Scholar] [CrossRef]
  80. Yanovski, S.Z.; Yanovski, J.A. Long-term drug treatment for obesity: A systematic and clinical review. JAMA 2014, 311, 74–86. [Google Scholar] [CrossRef]
  81. Sweeting, A.N.; Tabet, E.; Caterson, I.D.; Markovic, T.P. Management of obesity and cardiometabolic risk—Role of phentermine/extended-release topiramate. Diabetes Metab. Syndr. Obes. 2018, 11, 683–695. [Google Scholar] [CrossRef]
  82. PubChem. Topiramate. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Topiramate (accessed on 27 February 2025).
  83. Is Topamax Addictive? Medical News Today. Available online: https://www.medicalnewstoday.com/articles/drugs-is-topamax-addictive#potential-for-misuse-or-abuse (accessed on 27 February 2025).
  84. Gadde, K.M.; Allison, D.B.; Ryan, D.H.; Peterson, C.A.; Troupin, B.; Schwiers, M.L.; Day, W.W. Effects of low-dose, controlled-release phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): A randomized, placebo-controlled, phase 3 trial. Lancet 2011, 377, 1341–1352. [Google Scholar] [CrossRef] [PubMed]
  85. Garvey, W.T.; Ryan, D.H.; Look, M.; Gadde, K.M.; Allison, D.B.; Peterson, C.A.; Schwiers, M.; Day, W.W.; Bowden, C.H. Two-year sustained weight loss and metabolic benefits with controlled-release phentermine/topiramate in obese and overweight adults (SEQUEL): A randomized, placebo-controlled, phase 3 extension study. Am. J. Clin. Nutr. 2012, 95, 297–308. [Google Scholar] [CrossRef] [PubMed]
  86. Allison, D.B.; Gadde, K.M.; Garvey, W.T.; Peterson, C.A.; Schwiers, M.L.; Najarian, T.; Tam, P.Y.; Troupin, B.; Day, W.W. Controlled-release phentermine/topiramate in severely obese adults: A randomized controlled trial (EQUIP). Obesity 2012, 20, 330–342. [Google Scholar] [CrossRef]
  87. Fidler, M.C.; Sanchez, M.; Raether, B.; Weissman, N.J.; Smith, S.R.; Shanahan, W.R.; Anderson, C.M.; for the BLOSSOM Clinical Trial Group. A one-year randomized trial of lorcaserin for weight loss in obese and overweight adults: The BLOSSOM trial. J. Clin. Endocrinol. Metab. 2011, 96, 3067–3077. [Google Scholar] [CrossRef]
  88. PubChem. Bupropion. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/bupropion (accessed on 27 February 2025).
  89. Huecker, M.R.; Smiley, A.; Saadabadi, A. Bupropion. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK470212/ (accessed on 27 February 2025).
  90. PubChem. Naltrexone. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Naltrexone (accessed on 27 February 2025).
  91. Singh, D.; Saadabadi, A. Naltrexone. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK534811/ (accessed on 27 February 2025).
  92. Greenway, F.L.; Fujioka, K.; Plodkowski, R.A.; Mudaliar, S.; Guttadauria, M.; Erickson, J.; Kim, D.D.; Dunayevich, E.; for the COR-I Study Group. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): A multicenter, randomized, double-blind, placebo-controlled, phase 3 trial. Lancet 2010, 376, 595–605. [Google Scholar] [CrossRef]
  93. Wadden, T.A.; Foreyt, J.P.; Foster, G.D.; Hill, J.O.; Klein, S.; O’Neil, P.M.; Perri, M.G.; Pi-Sunyer, F.X.; Rock, C.L.; Erickson, J.S.; et al. Weight loss with naltrexone SR/bupropion SR combination therapy as an adjunct to behavior modification: The COR-BMOD trial. Obesity 2011, 19, 110–120. [Google Scholar] [CrossRef]
  94. Nissen, S.E.; Wolski, K.E.; Prcela, L.; Wadden, T.A.; Buse, J.B.; Bakris, G.; Perez, A.; Smith, S.R. Effect of naltrexone-bupropion on major adverse cardiovascular events in overweight and obese patients with cardiovascular risk factors: A randomized clinical trial. JAMA 2016, 315, 990–1004. [Google Scholar] [CrossRef] [PubMed]
  95. Davidson, M.H.; Hauptman, J.; DiGirolamo, M.; Foreyt, J.P.; Halsted, C.H.; Heber, D.; Heimburger, D.C.; Lucas, C.P.; Robbins, D.C.; Chung, J.; et al. Weight control and risk factor reduction in obese subjects treated for 2 years with orlistat: A randomized controlled trial. JAMA 1999, 281, 235–242. [Google Scholar] [CrossRef] [PubMed]
  96. Torgerson, J.S.; Hauptman, J.; Boldrin, M.N.; Sjöström, L. XENical in the prevention of diabetes in obese subjects (XENDOS) study: A randomized study of orlistat as an adjunct to lifestyle changes for the prevention of type 2 diabetes in obese patients. Diabetes Care 2004, 27, 155–161. [Google Scholar] [CrossRef] [PubMed]
  97. Bansal, A.B.; Al Khalili, Y. Orlistat. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2020. Available online: https://www.ncbi.nlm.nih.gov/books/NBK542202/ (accessed on 27 February 2025).
  98. PubChem. Orlistat. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Orlistat (accessed on 27 February 2025).
  99. U.S. Food & Drug Administration FDA. Orlistat (Marketed as Alli and Xenical) Information. Available online: https://www.fda.gov/drugs/postmarket-drug-safety-information-patients-and-providers/orlistat-marketed-alli-and-xenical-information (accessed on 27 February 2025).
  100. Sjöström, L.; Rissanen, A.; Andersen, T.; Boldrin, M.; Golay, A.; Koppeschaar, H.P.F.; Krempf, M.; for the European Orlistat Obesity Study Group. Randomized placebo-controlled trial of orlistat for weight loss and prevention of weight regain in obese patients. Lancet 1998, 352, 167–172. [Google Scholar] [CrossRef]
  101. Sweeting, A.N.; Hocking, S.L.; Markovic, T.P. Pharmacotherapy for the treatment of obesity. Mol. Cell. Endocrinol. 2015, 418, 173–183. [Google Scholar] [CrossRef]
  102. Zhou, J.; Massey, S.; Story, D.; Li, L. Metformin: An old drug with new applications. Int. J. Mol. Sci. 2018, 19, 2863. [Google Scholar] [CrossRef]
  103. Salpeter, S.R.; Buckley, N.S.; Kahn, J.A.; Salpeter, E.E. Meta-analysis: Metformin treatment in persons at risk for diabetes mellitus. Am. J. Med. 2008, 121, 149–157. [Google Scholar] [CrossRef]
  104. Diabetes Prevention Program Research Group. Long-term safety, tolerability, and weight loss associated with metformin in the Diabetes Prevention Program Outcomes Study. Diabetes Care 2012, 35, 731–737. [Google Scholar] [CrossRef]
  105. Metformin: History and Mechanism of Action. LGC Standards 2020. Available online: https://www.lgcstandards.com/GB/en/Resources/Articles/Pharma-roots-Metformin (accessed on 27 February 2025).
  106. Corcoran, C.; Jacobs, T.F. Metformin. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK518983/ (accessed on 27 February 2025).
  107. Knowler, W.C.; Barrett-Connor, E.; Fowler, S.E.; Hamman, R.F.; Lachin, J.M.; Walker, E.A.; Nathan, D.M.; Bennett, P.H.; Brown, S.A.; Goldberg, R.; et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N. Engl. J. Med. 2002, 346, 393–403. [Google Scholar] [CrossRef]
  108. Cusi, K.; DeFronzo, R.A. Metformin: A review of its metabolic effects. Diabetes Rev. 1998, 6, 89–131. [Google Scholar]
  109. Naderpoor, N.; Shorakae, S.; de Courten, B.; Misso, M.L.; Moran, L.J.; Teede, H.J. Metformin and lifestyle modification in polycystic ovary syndrome: Systematic review and meta-analysis. Hum. Reprod. Update 2015, 21, 560–574. [Google Scholar] [CrossRef] [PubMed]
  110. Gonzalez-Ortiz, M.; Martinez-Abundis, E.; Robles-Cervantes, J.A.; Ramos-Zavala, M.G.; Barraza-Salas, M. Effect of metformin on insulin sensitivity in obese nondiabetic patients: A double-blind placebo-controlled study. Metabolism 1999, 48, 586–589. [Google Scholar]
  111. Malin, S.K.; Kashyap, S.R. Effects of metformin on weight loss: Potential mechanisms. Curr. Opin. Endocrinol. Diabetes Obes. 2014, 21, 323–329. [Google Scholar] [CrossRef] [PubMed]
  112. Frias, J.P.; Deenadayalan, S.; Erichsen, L.; Jodar, E.; Lingvay, I.; Macura, S.; Mathieu, C.; Bajaj, H.S.; Goland, R.S.; Kalra, S.; et al. Efficacy and safety of co-administered once-weekly cagrilintide 2.4 mg with once-weekly semaglutide 2.4 mg in type 2 diabetes: A multicentre, randomised, double-blind, active-controlled, phase 2 trial. Lancet 2023, 402, 720–730. [Google Scholar] [CrossRef]
  113. Zhang, Y.; Duan, L.; Liu, T.; Zhang, H.; Wang, Y.; Li, Z.; Wang, X.; Wang, S.; Zhou, F.; Chen, R.; et al. Cagrilintide: A novel long-acting amylin analog for weight loss therapy. J. Med. Chem. 2021, 64, 1581–1589. [Google Scholar] [CrossRef]
  114. D’Ascanio, A.M.; Mullally, J.A.; Frishman, W.H. Cagrilintide: A long-acting amylin analog for the treatment of obesity. Cardiol. Rev. 2024, 32, 83–90. [Google Scholar] [CrossRef]
  115. Yao, H.; Zhang, A.; Li, D.; Liu, Q.; Li, X.; Li, Y.; Wang, Y.; Chen, J.; Hu, Z.; Wang, F.; et al. Comparative effectiveness of GLP-1 receptor agonists on glycaemic control, body weight, and lipid profile for type 2 diabetes: Systematic review and network meta-analysis. BMJ 2024, 384, e076410. [Google Scholar] [CrossRef]
  116. Enebo, L.B.; Berthelsen, K.K.; Kankam, M.; Lund, M.T.; Rubino, D.M.; Satylganova, A.; Lau, D.C.W. Safety, tolerability, pharmacokinetics, and pharmacodynamics of concomitant administration of multiple doses of cagrilintide with semaglutide 2.4 mg for weight management: A randomised, controlled, phase 1b trial. Lancet 2021, 397, 1736–1748. [Google Scholar] [CrossRef]
  117. Lau, D.C.W.; Erichsen, L.; Francisco, A.M.; Satylganova, A.; le Roux, C.W.; McGowan, B.; Pedersen, S.D.; Wharton, S.; Davies, M.J.; Zacho, J.; et al. Once-weekly cagrilintide for weight management in people with overweight and obesity: A multicentre, randomised, double-blind, placebo-controlled and active-controlled, dose-finding phase 2 trial. Lancet 2021, 398, 2160–2172. [Google Scholar] [CrossRef]
  118. Walpurgis, K.; Thomas, A.; Dellanna, F.; Schänzer, W.; Thevis, M. Detection of the human anti-ActRII antibody bimagrumab in serum by means of affinity purification, tryptic digestion, and LC-HRMS. Proteomics Clin. Appl. 2018, 12, e1700120. [Google Scholar] [CrossRef] [PubMed]
  119. Heymsfield, S.B.; Coleman, L.A.; Miller, R.; Rooks, D.S.; Laurent, D.; Petricoul, O.; Praestgaard, J.; Roubenoff, R.; Polidori, D.; MacDougall, D.; et al. Effect of bimagrumab vs placebo on body fat mass among adults with type 2 diabetes and obesity: A phase 2 randomized clinical trial. JAMA Netw. Open 2021, 4, e2033457. [Google Scholar] [CrossRef] [PubMed]
  120. Monoclonal antibodies. In LiverTox: Clinical and Research Information on Drug-Induced Liver Injury; National Institute of Diabetes and Digestive and Kidney Diseases: Bethesda, MD, USA, 2024. Available online: https://pubmed.ncbi.nlm.nih.gov/31644151/ (accessed on 27 February 2025).
  121. Nunn, E.; Jaiswal, N.; Gavin, M.; Del Rivero, J.; Gavini, C.K.; Altarejos, J.Y.; Breen, D.M.; Peltekian, K.M.; Heymsfield, S.B.; He, J.; et al. Antibody blockade of activin type II receptors preserves skeletal muscle mass and enhances fat loss during GLP-1 receptor agonism. Mol. Metab. 2024, 80, 101880. [Google Scholar] [CrossRef]
  122. Rooks, D.; Petricoul, O.; Praestgaard, J.; Bartlett, M.; Laurent, D.; Roubenoff, R. Safety and pharmacokinetics of bimagrumab in healthy older and obese adults with body composition changes in the older cohort. J. Cachexia Sarcopenia Muscle 2020, 11, 1525–1534. [Google Scholar] [CrossRef] [PubMed]
  123. Tchang, B.G.; Abel, B.; Zecca, C.; Shukla, A.P.; Aronne, L.J. An up-to-date evaluation of lorcaserin hydrochloride for the treatment of obesity. Expert Opin. Pharmacother. 2020, 21, 21–28. [Google Scholar] [CrossRef] [PubMed]
  124. Stamm, S.; Gruber, S.B.; Rabchevsky, A.G.; Emeson, R.B. The activity of the serotonin receptor 2C is regulated by alternative splicing. Hum. Genet. 2017, 136, 1079–1091. [Google Scholar] [CrossRef]
  125. U.S. FDA. Safety Clinical Trial Shows Possible Increased Risk of Cancer with Weight-Loss Medicine Belviq, Belviq XR (lorcaserin). Available online: https://www.fda.gov/drugs/fda-drug-safety-podcasts/safety-clinical-trial-shows-possible-increased-risk-cancer-weight-loss-medicine-belviq-belviq-xr (accessed on 27 February 2025).
  126. U.S. FDA. FDA Requests Withdrawal of Weight-Loss Drug Belviq, Belviq XR (lorcaserin) from the Market. Available online: https://www.fda.gov/drugs/drug-safety-and-availability/fda-requests-withdrawal-weight-loss-drug-belviq-belviq-xr-lorcaserin-market (accessed on 27 February 2025).
  127. Harvard Medical School. How Artificial Intelligence Is Disrupting Medicine & What It Means for Physicians. Available online: https://postgraduateeducation.hms.harvard.edu/trends-medicine/how-artificial-intelligence-disrupting-medicine-what-means-physicians (accessed on 27 February 2025).
  128. Chew, H.S.J. The use of artificial intelligence-based conversational agents (chatbots) for weight loss: Scoping review and practical recommendations. JMIR Med. Inform. 2022, 10, e32578. [Google Scholar] [CrossRef]
  129. Oh, Y.J.; Zhang, J.; Fang, M.L.; Fukuoka, Y. A systematic review of artificial intelligence chatbots for promoting physical activity, healthy diet, and weight loss. Int. J. Behav. Nutr. Phys. Act. 2021, 18, 160. [Google Scholar] [CrossRef]
  130. Chu, Y.T.; Huang, R.Y.; Chen, T.T.; Lee, Y.T.; Lee, Y.C.; Kuo, P.H.; Hsu, C.Y. Effect of health literacy and shared decision-making on weight-loss plan choice via an AI robot intervention. Digit. Health 2022, 8, 20552076221136372. [Google Scholar] [CrossRef]
  131. Fernandes, G.J.; Choi, A.; Schauer, J.M.; Pfammatter, A.F.; Spring, B.J.; Darwiche, A.; Alshurafa, N.I. An explainable artificial intelligence software tool for weight management experts (PRIMO): Mixed methods study. J. Med. Internet Res. 2023, 25, e42047. [Google Scholar] [CrossRef]
  132. Foster, G.D.; Wyatt, H.R.; Hill, J.O.; McGuckin, B.G.; Brill, C.; Mohammed, B.S.; Szapary, P.O.; Rader, D.J.; Edman, J.S.; Klein, S.; et al. Weight and metabolic outcomes after 2 years on a low-carbohydrate versus low-fat diet: A randomized trial. Ann. Intern. Med. 2010, 153, 147–157. [Google Scholar] [CrossRef] [PubMed]
  133. Wadden, T.A.; Walsh, O.A.; Berkowitz, R.I.; Leonard, S.M.; Swantek, J.S.; Chao, A.M.; Bakizada, Z.M.; Williams, N.N.; Alamuddin, N.; Dempsey, M.C.; et al. Intensive behavioral therapy for obesity combined with liraglutide 3.0 mg: A randomized controlled trial. Obesity 2019, 27, 75–86. [Google Scholar] [CrossRef] [PubMed]
  134. Venditti, E.M.; Wylie-Rosett, J.; Delahanty, L.M.; Mele, L.; Hoskin, M.A.; Edelstein, S.L.; the Diabetes Prevention Program Research Group. Short and long-term lifestyle coaching approaches used to address diverse participant barriers to weight loss and physical activity adherence. Int. J. Behav. Nutr. Phys. Act. 2014, 11, 16. [Google Scholar] [CrossRef] [PubMed]
  135. Leung, A.W.Y.; Chan, R.S.M.; Sea, M.M.M.; Woo, J. Psychological factors of long-term dietary and physical activity adherence among Chinese adults with overweight and obesity. Nutrients 2020, 12, 1379. [Google Scholar] [CrossRef] [PubMed]
  136. Yackobovitch-Gavan, M.; Steinberg, D.M.; Endevelt, R.; Benyamini, Y. Factors associated with dropout in a group weight-loss programme: A longitudinal investigation. J. Hum. Nutr. Diet. 2015, 28 (Suppl. S2), 33–40. [Google Scholar] [CrossRef]
  137. Colombo, O.; Ferretti, V.V.; Ferraris, C.; Trentani, C.; Vinai, P.; Villani, S.; Tagliabue, A. Is drop-out from obesity treatment a predictable and preventable event? Nutr. J. 2014, 13, 13. [Google Scholar] [CrossRef]
  138. Masood, W.; Annamaraju, P.; Khan Suheb, M.Z.; Uppaluri, K.R. Ketogenic Diet. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://pubmed.ncbi.nlm.nih.gov/29763005/ (accessed on 1 April 2025).
  139. Stubbs, B.J.; Cox, P.J.; Evans, R.D.; Cyranka, M.; Clarke, K.; de Wet, H. A ketone ester drink lowers human ghrelin and appetite. Obesity 2018, 26, 269–273. [Google Scholar] [CrossRef]
  140. Sumithran, P.; Prendergast, L.A.; Delbridge, E.; Purcell, K.; Shulkes, A.; Kriketos, A.; Proietto, J. Ketosis and appetite-mediating nutrients and hormones after weight loss. Eur. J. Clin. Nutr. 2013, 67, 759–764. [Google Scholar] [CrossRef]
  141. Johnstone, A.M.; Horgan, G.W.; Murison, S.D.; Bremner, D.M.; Lobley, G.E. Effects of a high-protein ketogenic diet on hunger, appetite, and weight loss in obese men feeding ad libitum. Am. J. Clin. Nutr. 2008, 87, 44–55. [Google Scholar] [CrossRef]
  142. Dashti, H.M.; Mathew, T.C.; Khadada, M.; Al-Mousawi, M.; Talib, H.; Asfar, S.K.; Behbahani, A.I.; Al-Zaid, N.S. Beneficial effects of ketogenic diet in obese diabetic subjects. Mol. Cell. Biochem. 2007, 302, 249–256. [Google Scholar] [CrossRef]
  143. Brinkworth, G.D.; Noakes, M.; Buckley, J.D.; Keogh, J.B.; Clifton, P.M. Long-term effects of a very-low-carbohydrate weight loss diet compared with an isocaloric low-fat diet after 12 months. Am. J. Clin. Nutr. 2009, 90, 23–32. [Google Scholar] [CrossRef] [PubMed]
  144. Moreno, B.; Bellido, D.; Sajoux, I.; Goday, A.; Saavedra, D.; Crujeiras, A.B.; Casanueva, F.F. Comparison of a very low-calorie ketogenic diet with a standard low-calorie diet in the treatment of obesity. Endocrine 2014, 47, 793–805. [Google Scholar] [CrossRef] [PubMed]
  145. Moreno, B.; Crujeiras, A.B.; Bellido, D.; Sajoux, I.; Casanueva, F.F. Obesity treatment by very low-calorie ketogenic diet at two years: Reduction in visceral fat and on the burden of disease. Endocrine 2016, 54, 681–690. [Google Scholar] [CrossRef] [PubMed]
  146. Kang, H.C.; Chung, D.E.; Kim, D.W.; Kim, H.D. Early- and late-onset complications of the ketogenic diet for intractable epilepsy. Epilepsia 2004, 45, 1116–1123. [Google Scholar] [CrossRef] [PubMed]
  147. Calderon, G.; Gonzalez-Izundegui, D.; Shan, K.L.; Campos, A.; Breen-Lyles, M.; Guerrero, A.; Acosta, A.; Nuttall, F.Q.; Heymsfield, S.B.; Kushner, R.F.; et al. Effectiveness of anti-obesity medications approved for long-term use in a multidisciplinary weight management program. Int. J. Obes. 2022, 46, 555–563. [Google Scholar] [CrossRef]
  148. Finer, N.; James, W.P.T.; Kopelman, P.G.; Lean, M.E.J.; Williams, G. One-year treatment of obesity: A randomized, double-blind, placebo-controlled, multicentre study of orlistat. Int. J. Obes. Relat. Metab. Disord. 2000, 24, 306–313. [Google Scholar] [CrossRef] [PubMed]
  149. Garvey, W.T.; Batterham, R.L.; Bhatta, M.; Busetto, L.; Frias, J.P.; Jódar, E.; Lau, D.C.W.; Halpern, A.; Kyle, T.K.; Yu, A.P.; et al. Two-year effects of semaglutide in adults with overweight or obesity: The STEP 5 trial. Nat. Med. 2022, 28, 2083–2091. [Google Scholar] [CrossRef] [PubMed]
  150. Lewis, K.H.; Fischer, H.; Ard, J.; Barton, L.; Bessesen, D.H.; Daley, M.F.; Desai, J.; McDermott, M.; Phimphasone-Brady, P.; Powers, J.D.; et al. Safety and effectiveness of longer-term phentermine use. Obesity 2019, 27, 591–602. [Google Scholar] [CrossRef]
  151. Poulton, A.S.; Hibbert, E.J.; Champion, B.L.; Connor, J.P.; Knight, C.; Winstock, A.R.; Nassar, N.; Smith, C.; Shum, D.H.K.; Schofield, P.R.; et al. Piloting a new approach to the treatment of obesity using dexamphetamine. Front. Endocrinol. 2015, 6, 14. [Google Scholar] [CrossRef]
  152. Müller, T.D.; Blüher, M.; Tschöp, M.H.; DiMarchi, R.D. Anti-obesity drug discovery: Advances and challenges. Nat. Rev. Drug Discov. 2022, 21, 201–223. [Google Scholar] [CrossRef]
  153. Seifarth, C.; Schehler, B.; Schneider, H.J. Effectiveness of metformin on weight loss in non-diabetic individuals with obesity. Exp. Clin. Endocrinol. Diabetes 2013, 121, 27–31. [Google Scholar] [CrossRef] [PubMed]
  154. Gasoyan, H.; Pfoh, E.R.; Schulte, R.; Dalal, M.R.; Aminian, A.; Arterburn, D.E.; Goodpaster, B.H.; Khandelwal, N.; Lewis, K.H.; Horberg, M.A.; et al. Early- and later-stage persistence with antiobesity medications: A retrospective cohort study. Obesity 2024, 32, 486–493. [Google Scholar] [CrossRef] [PubMed]
  155. Enright, C.; Thomas, E.; Saxon, D.R. An updated approach to antiobesity pharmacotherapy: Moving beyond the 5% weight loss goal. J. Endocr. Soc. 2023, 7, bvac195. [Google Scholar] [CrossRef]
  156. Topart, P. Obesity surgery: Which procedure should we choose and why? J. Visc. Surg. 2023, 160, S30–S37. [Google Scholar] [CrossRef] [PubMed]
  157. Sjöström, L. Review of the key results from the Swedish Obese Subjects (SOS) trial—A prospective controlled intervention study of bariatric surgery. J. Intern. Med. 2013, 273, 219–234. [Google Scholar] [CrossRef]
  158. O’Brien, P.E.; Hindle, A.; Brennan, L.; Skinner, S.; Burton, P.; Smith, A.; Crosthwaite, G.; Brown, W. Long-term outcomes after bariatric surgery: A systematic review and meta-analysis of weight loss at 10 or more years. Obes. Surg. 2019, 29, 3–14. [Google Scholar] [CrossRef]
  159. Collazo-Clavell, M.L.; Shah, M. Common and rare complications of bariatric surgery. Endocrinol. Metab. Clin. N. Am. 2020, 49, 329–346. [Google Scholar] [CrossRef] [PubMed]
  160. Kim, J.C.; Kim, M.G.; Park, J.K.; Lee, J.H.; Hwang, Y.J.; Lim, H.S. Outcomes and adverse events after bariatric surgery: An updated systematic review and meta-analysis, 2013–2023. J. Metab. Bariatr. Surg. 2023, 12, 76–88. [Google Scholar] [CrossRef]
  161. Goldfeder, L.B.; Ren, C.J.; Gill, J.R. Fatal complications of bariatric surgery. Obes. Surg. 2006, 16, 1050–1056. [Google Scholar] [CrossRef]
  162. Doble, B.; Wordsworth, S.; Rogers, C.A.; Reeves, B.C.; Blazeby, J.M. What are the real procedural costs of bariatric surgery? Obes. Surg. 2017, 27, 2179–2192. [Google Scholar] [CrossRef]
  163. Fuller, N.R.; Pearson, S.; Lau, N.S.; Wiggins, B.; Shrestha, R.; Rao, A.; Caterson, I.D. An intragastric balloon in the treatment of obese individuals with metabolic syndrome: A randomized controlled study. Obesity 2013, 21, 1561–1570. [Google Scholar] [CrossRef] [PubMed]
  164. Lopez-Nava, G.; Asokkumar, R.; Bautista-Castaño, I.; Al-Mrabeh, A.; Delhom, E.; Fernández-Corbelle, J.P.; Espinet-Coll, E. Endoscopic sleeve gastroplasty, laparoscopic sleeve gastrectomy, and laparoscopic greater curve plication: Do they differ at 2 years? Endoscopy 2021, 53, 235–243. [Google Scholar] [CrossRef] [PubMed]
  165. Sullivan, S.; Swain, J.M.; Woodman, G.; Anton, T.; Gonzalez-Campoy, J.M.; Mehta, A.; Al-Amin, A.; Sheikh, E.; Chao, C.; Zhang, X.; et al. Randomized sham-controlled trial evaluating efficacy and safety of endoscopic gastric plication for primary obesity: The ESSENTIAL trial. Obesity 2017, 25, 294–301. [Google Scholar] [CrossRef]
  166. Sullivan, S.; Stein, R.; Jonnalagadda, S.; Mullady, D.; Kantsevoy, S.; Curvers, W.; Edmundowicz, S.A.; Abu Dayyeh, B.K.; Thompson, C.C.; Shah, R.J.; et al. Aspiration therapy leads to weight loss in obese subjects: A pilot study. Gastroenterology 2013, 145, 1245–1252.e5. [Google Scholar] [CrossRef] [PubMed]
  167. Jirapinyo, P.; Hadefi, A.; Thompson, C.C. ASGE-ESGE guideline on primary endoscopic bariatric and metabolic therapies for adults with obesity. Endoscopy 2024, 56, 437–456. [Google Scholar] [CrossRef]
  168. Abu Dayyeh, B.K.; Acosta, A.; Camilleri, M.; Mundi, M.S.; Rajan, E.; Thompson, C.C.; Sharma, A.M.; Edmundowicz, S.A.; Ginsberg, G.G.; Rothstein, R.I.; et al. Endoscopic sleeve gastroplasty alters gastric physiology and induces loss of body weight in obese individuals. Clin. Gastroenterol. Hepatol. 2017, 15, 37–43.e1. [Google Scholar] [CrossRef] [PubMed]
  169. Ponce, J.; Quebbemann, B.B.; Patterson, E.J.; Sharma, A.M.; Jirapinyo, P.; Kim, H.; Yellumahanthi, K.; Barham, H.P.; Clements, R.H.; DeLuca, L.; et al. Prospective, randomized, multicenter study evaluating safety and efficacy of intragastric dual-balloon in obesity. Surg. Obes. Relat. Dis. 2013, 9, 290–295. [Google Scholar] [CrossRef]
  170. Saumoy, M.; Gandhi, D.; Buller, S.; Silver, E.; Shah, S.; Landau, C.; Khorsandi, P.; Chokshi, R.; Carr-Locke, D.L.; Jirapinyo, P.; et al. Cost-effectiveness of endoscopic, surgical and pharmacological obesity therapies. Gut 2023, 72, 2250–2259. [Google Scholar] [CrossRef] [PubMed]
  171. Khera, R.; Murad, M.H.; Chandar, A.K.; Dulai, P.S.; Wang, Z.; Prokop, L.J.; Loomba, R.; Camilleri, M.; Singh, S.; Mullen, K.D.; et al. Association of pharmacological treatments for obesity with weight loss and adverse events: A systematic review and meta-analysis. JAMA 2016, 315, 2424–2434. [Google Scholar] [CrossRef]
  172. Kim, K.K.; Cho, H.J.; Kang, H.C.; Youn, B.B.; Lee, K.R. Effects on weight reduction and safety of short-term phentermine administration in Korean obese people. Yonsei Med. J. 2006, 47, 614–625. [Google Scholar] [CrossRef]
  173. Padwal, R.; Li, S.K.; Lau, D.C. Long-term pharmacotherapy for obesity and overweight. Cochrane Database Syst. Rev. 2004, 2003, CD004094. [Google Scholar] [CrossRef] [PubMed]
  174. Lenharo, M. How anti-obesity drugs cause nausea: Finding offers hope for better drugs. Nature 2024, 631, 493–494. [Google Scholar] [CrossRef] [PubMed]
  175. Apovian, C.M.; Aronne, L.; Rubino, D.; Still, C.; Wyatt, H.; Burns, C.; Kim, D.; Dunayevich, E.; Ascher, S.; Hall, G.; et al. A randomized, phase 3 trial of naltrexone SR/bupropion SR on weight and obesity-related risk factors (COR-II). Obesity 2013, 21, 935–943. [Google Scholar] [CrossRef] [PubMed]
  176. O’Neil, P.M.; Birkenfeld, A.L.; McGowan, B.; Mosenzon, O.; Pedersen, S.D.; Wharton, S.; Carson, C.G.; Jepsen, C.H.; Kabisch, M.; Wilding, J.P.H.; et al. Efficacy and safety of semaglutide compared with liraglutide and placebo for weight loss in patients with obesity: A randomised, double-blind, placebo and active controlled, dose-ranging, phase 2 trial. Lancet 2018, 392, 637–649. [Google Scholar] [CrossRef]
  177. PubMed. Post-Marketing Safety of Anti-Obesity Medications: Analysis of the FDA Adverse Event Reporting System (FAERS). Available online: https://pubmed.ncbi.nlm.nih.gov/34564826/ (accessed on 27 February 2025).
  178. PubMed. Safety and Efficacy of Long-Term Phentermine Use in Obesity Management. Available online: https://pubmed.ncbi.nlm.nih.gov/27478924/ (accessed on 27 February 2025).
  179. U.S. FDA Guidance Document. Developing Products for Weight Management. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/developing-products-weight-management-revision-1 (accessed on 27 February 2025).
  180. EMA. Guideline on Clinical Evaluation of Medicinal Products Used in Weight Management. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-clinical-evaluation-medicinal-products-used-weight-management-revision-1_en.pdf (accessed on 27 February 2025).
  181. ICER. Policy Recommendations for Obesity Management. Available online: https://icer.org/wp-content/uploads/2022/10/ICER_Obesity_Policy_Recommendations_102022.pdf (accessed on 27 February 2025).
  182. ASOI. Pharmacotherapy in Obesity Management. Available online: https://asoi.info/wp-content/uploads/2022/10/11-IR-Pharmacotherapy-in-Obesity-Management-FINAL.pdf (accessed on 27 February 2025).
Figure 1. Schematic diagram showing the approach to treating obesity. Created with BioRender software.
Figure 1. Schematic diagram showing the approach to treating obesity. Created with BioRender software.
Diseases 13 00129 g001
Figure 2. Timeline of FDA approvals for weight loss parenteral medications.
Figure 2. Timeline of FDA approvals for weight loss parenteral medications.
Diseases 13 00129 g002
Table 1. Comparison of weight management modalities: mechanisms, efficacy, and considerations.
Table 1. Comparison of weight management modalities: mechanisms, efficacy, and considerations.
Modality TypeDescriptionEfficacy and TimeframePatient SuitabilitySide Effects and AdherenceCost (Approx.)References
Lifestyle interventionsLow-calorie diet (500–750 kcal/day), aerobic and resistance training, behavior therapy6–11% weight loss in 1 yearBMI 25–29.9 (no comorbidities) or BMI ≥ 30 with comorbiditiesNon-adherence: 21–60%, barriers include time and motivationN/A[77,132,133,134,135,136,137]
Dietary readjustment—ketogenic dietHigh-fat, low-carbohydrate diet shifts metabolism toward fat oxidation and ketone production; ketones suppress ghrelin, reduce appetite, and promote weight loss.14.5–30 kg weight loss over 1 yearNo contraindications (e.g., pancreatitis, hepatic failure, fat metabolism or carnitine disorders, porphyrias). Use with caution in diabetics on insulin/oral hypoglycemics due to hypoglycemia risk.Hypertriglyceridemia, transient hyperuricemia, hypercholesterolemia, renal stones, iron-deficiency anemiaN/A[138,139,140,141,142,143,144,145,146]
PharmacotherapyGLP-1 agonists (Semaglutide, Liraglutide), lipase inhibitors (Orlistat), MC4 agonists, appetite suppressants, insulin sensitizers4–18.6% weight loss in 6–12 monthsBMI ≥ 30 or BMI ≥ 27 with comorbiditiesAdherence: 10–40%, side effects: nausea, headache, constipationUSD 11–USD 1576/month[77,147,148,149,150,151,152,153,154,155]
Bariatric surgeryGastric bypass, sleeve gastrectomy, gastric banding, duodenal switch17–28% weight loss over 1–2 years
12–25% weight loss over 3–10 years
18% weight loss sustained over 20 years
BMI ≥ 40 or BMI ≥ 35 with comorbiditiesSide effects: nutritional deficiency, reflux, obstruction, bone lossUSD 7423–USD 33,541[77,156,157,158,159,160,161,162]
Endoscopic proceduresIntragastric balloons, endoscopic sleeve gastroplasty (ESG), POSE, AspireAssist4.95–21% weight loss in 1 yearBMI ≥ 30 or ≥27 with comorbiditiesSide effects: nausea, vomiting, heartburn, pulmonary risksUSD 4105–USD 11,411/QALY[77,163,164,165,166,167,168,169,170]
Combination/Adjunctive StrategySimultaneous or sequential use of two or more therapies (e.g., lifestyle + pharmacotherapy, endoscopy + behavioral intervention) to improve long-term outcomes and adherence6–13% total weight loss, depending on combination and adherence
Individuals with insufficient response to monotherapy or those at high risk of dropout.
BMI ≥ 30 with comorbidities
Adherence improves with behavioral support; side effects depend on pharmacologic or surgical componentsVariable depending on treatment mix; often higher than monotherapy alone[132,133,147,149]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Al Lawati, A.; Alhabsi, A.; Rahul, R.; Savino, M.-L.; Alwahaibi, H.; Das, S.; Al Lawati, H. Current and Emerging Parenteral and Peroral Medications for Weight Loss: A Narrative Review. Diseases 2025, 13, 129. https://doi.org/10.3390/diseases13050129

AMA Style

Al Lawati A, Alhabsi A, Rahul R, Savino M-L, Alwahaibi H, Das S, Al Lawati H. Current and Emerging Parenteral and Peroral Medications for Weight Loss: A Narrative Review. Diseases. 2025; 13(5):129. https://doi.org/10.3390/diseases13050129

Chicago/Turabian Style

Al Lawati, Abdullah, Ayman Alhabsi, Rhieya Rahul, Maria-Luisa Savino, Hamed Alwahaibi, Srijit Das, and Hanan Al Lawati. 2025. "Current and Emerging Parenteral and Peroral Medications for Weight Loss: A Narrative Review" Diseases 13, no. 5: 129. https://doi.org/10.3390/diseases13050129

APA Style

Al Lawati, A., Alhabsi, A., Rahul, R., Savino, M.-L., Alwahaibi, H., Das, S., & Al Lawati, H. (2025). Current and Emerging Parenteral and Peroral Medications for Weight Loss: A Narrative Review. Diseases, 13(5), 129. https://doi.org/10.3390/diseases13050129

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

Article Metrics

Back to TopTop