Introduction


Since the mid-twentieth century, the World Health Organization (WHO) has recognized "obesity" as a stand-alone, chronic, non-communicable and preventable disease that remains a major, global public health challenge 1, 2. Today, more than a billion individuals, both adults and children, worldwide are obese 3, with 110 million obese people in the US alone 4. Without intervention, nearly half the adults in the US are expected to be obese by 2030 4.

Objective criteria for diagnosis of clinical obesity and pre-obesity exist 5 , which provide a framework for clinical decision-making and prioritization of therapeutic interventions (Figure 1).

 
 
 
 
 
 
 
 
 
 
 
 
Diagnosis Normal Pre-clinical
Obesity
Clinical
Obesity
Excess adiposity? No Yes Yes
Muscle Mass Normal - High Normal - Low Normal - Low
Evidence of organ dysfunction due to adiposity? No No Yes
Negative impact on day-to-day activities? No No Yes
Higher health risk No Yes Yes

 

Figure 1: Overview of diagnostic criteria for different types of obesity. The state of excess adiposity leads to a diagnosis of obesity. The type of obesity is dependent on whether the excess adiposity negatively impacts organ structure and function. The risk of developing other co-morbid non-communicable conditions is increased for individuals with pre-clinical and clinical obesity. Generally, obesity is "a condition characterized by excess adiposity, with or without abnormal distribution or function of adipose tissue, and with causes that are multifactorial and still incompletely understood" 5.

Contents


1. Brief History of Early Weight-Loss Drugs in the US

The medicalization of obesity, which has been formerly underway since the mid twentieth century has been full of promise and demise. The first FDA approved drugs for weight management came in 1947 with Abbott Laboratories' Desoxyn and Endo Products' Hydrin 6. These anorectics were formulations of methamphetamine that served as pharmaceutical adjuncts to dietary management of obesity 6,7. While amphetamine-based anorectics were considered useful weight-loss drugs because they suppressed appetite, they carried a risk of addiction. Consequently, amphetamine analogues were developed with the intention of reducing this risk 6, 7. They were, however, indicated for short-term use which ultimately reduced their popularity 6. In the mid-1990s, dexfenfluramine was approved for long-term use in the treatment of obesity, but its dubious safety profile restricted its use, and it was eventually withdrawn from the US market 6.

Similarly, sibutramine (Meridia), a serotonin and dopamine reuptake inhibitor that works by inducing satiety and increasing energy expenditure 8, was approved in 1997 against a background of divided opinion on its safety profile because, while effective for weight-loss, it tended to increase blood pressure and pulse rate 6. At the request of the European Medical Agency, the SCOUT (Sibutramine Cardiovascular OUTcome Trial) study was undertaken to provide post-marketing assurance of cardiovascular safety for use of the drug in high-risk individuals. The drug was voluntarily withdrawn in 2010 9, 10 following an FDA request based on the increased risk of non-fatal myocardial infarction and non-fatal stroke in patients with preexisting cardiovascular disease identified via the SCOUT study, despite the drug not being indicated for such patients 8, 11.

More recently, lorcaserin (Belviq), a 5-hydroxytryptamine receptor agonist that functioned as an appetite suppressant 12, was approved in 2012 but was withdrawn in 2020, due to the increased risk of cancer based on emergent clinical data 13.

↑ Back to top

2. Biological Targets for Weight-Loss Drug Development

Anti-obesity drugs target either central nervous system pathways that regulate sensations of satiety and fullness, or peripheral modulators of digestion, metabolism and lipogenesis (Figure 2).

Figure 2: Simplified schematic overview of biological pathways targeted by weight-management drugs. Anorexigenic effects are mediated through stimulation of MC4R, which may be achieved via input signals, specifically insulin, amylin and leptin, from peripheral organs, including stomach, intestines and adipose tissue that act on POMC expressing neurons in the arcuate nucleus of the hypothalamus. Amylin also stimulates leptin signaling. Peptides AgRP and NPY are MC4R antagonists that are co-expressed in response to signals from elsewhere in the body that inform on energy stores. These signals are mediated through leptin and the so called "hunger hormone" ghrelin. Peripherally produced PYY also transmits a satiety signal to the brain by binding receptors on the vagus nerve. This signal prevents and stimulates NPY and POMC production by neurons in the arcuate nucleus respectively, resulting in orexigenic effects.  GLP1, glucagon-like-peptide-1; GLP1R, glucagon-like-peptide-1-receptor; GIP, glucose-dependent insulinotropic polypeptide; LEPR, leptin receptor; NPY, neuropeptide Y; PYY, peptide YY; POMC, proopiomelanocortin; PCSK1, Proprotein convertase subtilisin/kexin type 1; AgRP, Agouti-related protein; NPY, neuropeptide Y; αMSH, α-melanocyte stimulating hormone; MC4R, melanocortin receptor 4. Adapted from 14. Blue arrows are indicative of stimulation, while the red lines are indicative of inhibition.

↑ Back to top

3. US Weight Management Drug Development Pipeline

Presently, there are five patent-protected anti-obesity drugs approved for the US market (Table 1), and 149 novel drug candidates in clinical development either in the United States or globally 15. Across the various development stages, from Phase I to pre-registration, 101 different companies have contributed at least one novel drug candidate to the development pipeline, (Figure 3 a), indicative of the perceived opportunity that continues to exist in the medical management of obesity. Approximately 16% of drug candidates in the pipeline are being developed by either Novo Nordisk or Eli Lilly, underlining their status as leaders in the field and emphasising their commitment to the obesity market.

Biologics are generally well represented across Phase I to Phase III, with peptide-based drugs among the most numerically dominant at each stage (Figure 3 b). Twenty-seven small molecule drug candidates exist at Phase I, and at Phase II, small molecule drugs match the number of peptide-based drugs in development. Oligonucleotide drugs represent a new modality for treatment of obesity, and all candidates are currently in Phase I trials. Three monoclonal antibody drug candidates are also being developed, two of which (apitegromab and trevogrumab) are intended for use alongside marketed weight-loss drugs to preserve lean muscle mass during medicalised weight-loss.

Leveraging incretin biology for weight-loss remains popular, with ~35% of drug candidates exploiting this mechanism of action, (Figure 3 c). Amylin receptor biology is a mechanistic focus of ~7% of drug candidates, and ~3% of drugs in clinical development target both incretin and amylin receptor biology for weight-loss. Indicative of the innovation that exists within the pipeline, more than thirty mechanisms of action are represented by the ~33% of drug candidates with mechanism of action classified as "Other", (Figure 3 c).

In recognition of the need for alternatives to injectable drugs, ~44% of drug candidates are intended for oral delivery, either exclusively, or as an alternative to another mode of delivery, (Figure 3 d).

With ~7% of all drug candidates already in the later stages of development, (Phase III, pre-registration), (Figure 3 e; Table 2), it is likely that new weight-loss drugs will be marketed in the near-term future 

Table 1: Novel weight-loss drugs approved in the US
Tip: Click column headers to sort
← Scroll horizontally →
Generic Name Brand Name Company Name First Approval (Yr) Expected Expiry Label Indication Route of Administration Molecule Type Target Biology
tirzepatide Zepbound Eli Lilly and Co 2023 Jun-2039 Indicated in combination with a reduced-calorie diet and increased physical activity:
  • to reduce excess body weight and maintain weight reduction long term in adults with obesity or adults with overweight in the presence of at least one weight-related comorbid condition.
Subcutaneous Synthetic Peptide Incretin Biology
semaglutide Wegovy Novo Nordisk Inc 2021 Dec-2031 Indicated in combination with a reduced calorie diet and increased physical activity:
  • to reduce the risk of major adverse cardiovascular events (cardiovascular death, non-fatal myocardial infarction, or non-fatal stroke) in adults with established cardiovascular disease and either obesity or overweight.
  • to reduce excess body weight and maintain weight reduction long term in:
    • Adults and pediatric patients aged 12 years and older with obesity
    • Adults with overweight in the presence of at least one weight-related comorbid condition
Subcutaneous Recombinant Peptide Incretin Biology
setmelanotide Imcivree Rhythm Pharmaceuticals Inc 2020 Aug-2032 Indicated to reduce excess body weight and maintain weight reduction long term in adults and pediatric patients 2 years of age and older with syndromic or monogenic obesity due to:
  • Bardet-Biedl syndrome (BBS).
  • Pro-opiomelanocortin (POMC), proprotein convertase subtilisin/kexin type 1 (PCSK1), or leptin receptor (LEPR) deficiency as determined by an FDA-approved test demonstrating variants in POMC, PCSK1, or LEPR genes that are interpreted as pathogenic, likely pathogenic, or of uncertain significance (VUS).
Subcutaneous Synthetic Peptide Melanocortin Biology
(bupropion hydrochloride + naltrexone) Contrave Currax Pharmaceuticals LLC 2014 Feb-2029 Indicated as an adjunct to a reduced-calorie diet and increased physical activity for chronic weight management in adults with an initial body mass index (BMI) of:
  • 30 kg/m² or greater (obese) or
  • 27 kg/m² or greater (overweight) in the presence of at least one weight-related comorbidity
Oral Small Molecule Opioid Receptor Biology; Biology of Dopamine and Noradrenaline Transporter Inhibitors
(phentermine hydrochloride + topiramate) Qsymia Vivus LLC 2012 May-2029 Indicated in combination with a reduced-calorie diet and increased physical activity to reduce excess body weight and maintain weight reduction long term in:
  • Adults and pediatric patients aged 12 years and older with obesity
  • Adults with overweight in the presence of at least one weight-related comorbid condition
Oral Small Molecule Unknown

Navigation Tips: • First column and header row are frozen for easy reference • Scroll horizontally and vertically to view all data • Hover over rows to highlight them • Use compact mode to fit more content • Search to quickly find specific drugs • Hover over truncated text to see full content

 

Table 2: Late-stage anti-obesity drug development pipeline in the US
Tip: Click column headers to sort • Scroll horizontally and vertically
← Scroll horizontally →
Development Stage Drug Name Company Name Drug Geography Route of Administration Molecule Type Target Biology
Pre-Registration NN-9932 Novo Nordisk AS United States Oral Recombinant Peptide Incretin Biology
  (cagrilintide + semaglutide) Novo Nordisk AS China; Global Subcutaneous Recombinant Peptide; Synthetic Peptide Amylin Receptor Biology
  cagrilintide Novo Nordisk AS Global; Japan Oral; Subcutaneous Synthetic Peptide Amylin Receptor Biology
  ecnoglutide HK inno.N Corp Global Subcutaneous Biologic Incretin Biology
Phase III maridebart cafraglutide Amgen Inc Global Subcutaneous Fusion Protein Incretin Biology
  NN-9932 Novo Nordisk AS Asia-Pacific; China; EU; Global Oral Recombinant Peptide Incretin Biology
  orforglipron calcium Eli Lilly and Co China; EU; Global; Japan; United States Oral Small Molecule Incretin Biology
  retatrutide Eli Lilly and Co Global; India Subcutaneous Synthetic Peptide Incretin Biology
  semaglutide Chia Tai Tianqing Pharmaceutical Group Co Ltd Global Subcutaneous Recombinant Peptide Incretin Biology
  survodutide Boehringer Ingelheim International GmbH China; Global; Japan Subcutaneous Synthetic Peptide Incretin Biology

 

 

 

 

 

 

Figure 3: Overview of US and global anti-obesity drug development pipeline. The pipeline currently includes 149 drug candidates, (Phase I to Pre-registration inclusive) in development in the US or globally. (a) Companies with anti-obesity drug candidates in development. (b) Drug candidates in the various phases of development across various modalities. (c) Proportion of drugs in development at each development stage. (d) Biology targeted by mechanisms of action of drug candidates in development. "Multiple" refers to drug candidates that target more than one of the named biological targets, e.g. Incretin biology and amylin receptor biology. (e) Intended delivery modes for drugs candidates in development. Note that percentages in panels (c-e) are rounded to the nearest whole number.

↑ Back to top

4. Leveraging Incretin Hormone Biology for Drug Discovery and Development

Incretin hormones were an obvious starting point for development of weight-loss drugs. GIP and GLP-1 are hormones released after ingestion of food to control insulin secretion and to maintain glucose homeostasis, among other physiological effects 16, 17. Upon release, GLP-1 slows stomach emptying, stimulates insulin secretion in the pancreas, and via signalling in the brain, inhibits appetite and food intake. The "incretin effect" refers to the greater insulin secretion response stimulated by oral versus intravenous glucose delivery, even when identical blood glucose levels are achieved 18. This difference is attributed to the incretin hormones, GLP-1 and GIP, which are released in response to orally received, but not systemically delivered, glucose.

Noteworthy Successes

Foremost among the modern-day anti-obesity drugs are GLP-1 receptor agonists (GLP1RAs). These drugs mimic the binding of the naturally produced GLP-1 hormone to its cognate receptor after food intake, which has the effect of regulating glucose homeostasis, lipid metabolism, gastric emptying and feelings of satiety 19 (Figure 1).

Novo Nordisk has developed 20 and marketed two GLP1RAs, liraglutide (Saxenda) and semaglutide (Wegovy), both recombinant peptides, for treatment of obesity. (Semaglutide is also marketed as Ozempic for type 2 diabetes (T2D)). Liraglutide is taken by daily subcutaneous injection, while semaglutide is injected weekly 15. The intravenous half-life of native human GLP-1 is mere minutes 20. Consequently, enabling molecular engineering and chemistry was undertaken by Novo Nordisk to generate these GLP-1 analogues with a suitably long half-life and potency 20. Both drugs are intended for use as adjuncts alongside caloric restriction and increased physical activity for weight management in obese or overweight adults and children aged 12 to 17 years with at least one weight-related comorbidity, such as T2D, dyslipidemia or hypertension, 15, 21.

Novo Nordisk is currently progressing eleven drug candidates through clinical development, the most advanced of which are NN-9932 (semaglutide), CagriSema (cagrilintide + semaglutide), and cagrilintide, and are either in pre-registration or Phase III clinical trials (Table 2). Across Novo Nordisk's anti-obesity portfolio, at least half of all candidates are based on mechanisms of action that include GLP-1 receptor or GIP receptor agonism 15. Amylin receptor biology also features as a target for half of the emergent drug candidates 15.

Eli Lilly has developed tirzepatide, a novel, first-in-class, 15 synthetic peptide designed to be a dual agonist of GLP-1 and GIP 22. The drug is marketed as Zepbound for treatment of overweight/obese patients and Mounjaro for treatment of T2D. It is delivered weekly via subcutaneous injection. Eli Lilly sponsored SURMOUNT-5, (NCT05822830), a Phase IIIb, open label, multi-centre, randomised clinical trial to compare the efficacy and safety of tirzepatide (Zepbound) versus semaglutide (Wegovy) in overweight or obese adults and at least one additional named weight-related comorbidity that was not T2D 24. Greater weight loss and reduction in waist circumference was achieved with tirzepatide compared to semaglutide at 72 weeks 23.

Like Novo Nordisk, Eli Lilly also has a considerable portfolio of 11 novel drugs in development for treatment of obesity, five of which are either GLP1RAs or GIPRAs 15. The other assets are based on amylin receptor biology, activin receptor biology and peptide YY. Most products in this portfolio are peptides, though two small molecules and one monoclonal antibody are also being developed. Orforglipron calcium (licensed from Chugai Pharmaceutical Co. Ltd) and retatrutide are the most advanced and are in Phase III clinical trials (Table 2). At least three of the drug candidates in development are intended for oral delivery 15.

Noteworthy Failures

While Novo Nordisk and Eli Lilly have enjoyed considerable success with their incretin mimetics, Pfizer has not yet reaped the reward of its efforts in developing similar drugs for chronic weight management. Pfizer has unsuccessfully attempted to develop two GLP1RA drug candidates, lotiglipron (PF-07081532), co-developed with Heptares Therapeutics Ltd., and danuglipron (PF-06882961), both small molecules and new molecular entities intended for oral delivery for treatment of obesity. Lotiglipron was discontinued in 2023 owing to unfavourable pharmacokinetic drug-drug interaction data from two Phase I studies and to elevated transaminases indicative of liver dysfunction in a subset of patients 24.

Pfizer's refocused efforts on danuglipron also ended in a decision to discontinue its development in April 2025 following a review of all available clinical trial data, regulatory input and after a single asymptomatic clinical trial participant exhibited potential drug-induced liver injury that resolved after the drug was withdrawn 25. Despite these setbacks, Pfizer appears committed to the obesity market and continues to progress PF-07976016, a first-in-class, orally delivered small molecule GIP receptor antagonist for treatment of obesity, for which safety and efficacy is currently being evaluated in a dose-ranging Phase II study, (NCT06717425).

↑ Back to top

5. Targeting the Melanocortin System to Treat Hereditary Obesity

While obesity most often arises from lifestyle choices, particularly surrounding the balance of caloric intake and physical activity 26, 27, genetics may also play a role in a small subset of individuals. Loss-of-function mutations affecting genes in the leptin-melanocortin pathway, particularly MC4R, POMC, PCSK1 and LEPR, (Figure 2), predispose individuals to hyperphagia and severe early onset obesity 28 (Table 3).

The central melanocortin system is a neuro-immuno-endocrine hormone system that encompasses several central nervous system circuits 29 and controls various physiological functions, including food intake 29, 30. Melanocortins are a family of neuromodulatory peptides derived from the polypeptide, pro-opiomelanocortin (POMC), by tissue-specific cleavage 31, 32 through the action of prohormone convertases 31. Pro-peptide POMC expression is induced by binding of leptin to the LEPR on POMC neurons in the arcuate nucleus of the hypothalamus 28, 30. Proprotein convertase subtilisin/kexin type 1 (PCSK1), initiates cleavage of POMC into melanocortin family peptides, including α- and β- melanocyte stimulating hormones (αMSH, βMSH) 30. These melanocortin ligands activate the MC4R on neurons in the paraventricular nucleus of the hypothalamus 28, thereby suppressing appetite, promoting satiety and stimulating energy expenditure 33 (Figure 2).

Table 3: Selected genetic causes of obesity
Tip: Click column headers to sort
← Scroll horizontally →
Disease Gene Affected Prevalence Inheritance Pattern Age of Onset Reference
MC4R Deficiency MC4R (1-5)/10,000 Autosomal Dominant, Autosomal Recessive Infancy, Neonatal ORPHA:71529
POMC Deficiency POMC <1 /1,000,000 Autosomal Recessive Infancy, Neonatal ORPHA:71526
PCSK1 deficiency PCSK1 <1 /1,000,000 Autosomal Recessive Infancy, Neonatal ORPHA:71528
LEPR Deficiency LEPR Not given Autosomal Recessive Childhood, Infancy ORPHA:179494
Genetic non-syndromic obesity Not given Unknown Incidental Childhood, Infancy ORPHA:98267
Severe early-onset obesity-insulin resistance syndrome due to SH2B1 deficiency SH2B1 and 16p11.2 chromosomal deletions <1 /1,000,000 Autosomal Dominant Childhood ORPHA:329249
Bardet-Biedl Syndrome Many (1-2)/100,000 Autosomal recessive, Oligogenic Antenatal, Childhood, Infancy, Neonatal ORPHA:110
Alström Syndrome ALMS1 (1-9)/1,000,000 Autosomal recessive Adolescent, Adult, Childhood, Infancy, Neonatal ORPHA:64
Prader-Willi Syndrome Paternally derived 15q11-q13 deletion, maternal disomy or, very rarely, imprinting defects in the same region (1-9)/100,000 Variable Antenatal, Neonatal ORPHA:739

Navigation Tips: • First column and header row are frozen for easy reference • Scroll horizontally and vertically to view all data • Hover over rows to highlight them • Use compact mode to fit more content • Hover over truncated text to see full content

Setmelanotide, an MC4R agonist and first-in-class treatment for hereditary obesity

Setmelanotide, (Imcivree), is an MC4R agonist discovered by Ipsen SA and developed by Rhythm Pharmaceuticals that has been approved in the US since 2020 for treatment of obesity due to deficiencies of POMC, PCSK1 or LEPR confirmed by genetic testing 15 and since 2022 for syndromic obesity due to Bardet-Biedl Syndrome, a rare genetic ciliopathy 34. Its utility in treating a variety of other syndromic obesities is still being evaluated 15. In patients aged two years and older 35, Imcivree is delivered by daily subcutaneous injection 36.

Setmelanotide is a disulfide cyclic synthetic peptide, eight amino acids long, capable of crossing the blood-brain barrier 37 where at its site of action in the hypothalamus, it can activate several melanocortin receptors 37. It is a potent MC4R agonist at nanomolar concentrations, 38 and exhibits high affinity for the receptor. It is reasonably stable with an elimination half-life of approximately 11 hours 36. By agonising MC4R, it restores appetite control and increases energy expenditure 33 without directly treating the underlying genetic causes of obesity that occur upstream of MC4R activation. Setmelanotide was proven effective for weight loss in these genetically defined populations and is generally well tolerated 39.

Drug Development Pipeline for Genetic Causes of Obesity

Rhythm Pharmaceuticals is also developing investigational drugs, RM-718 and bivamelagon, both MC4R agonists for treatment of hypothalamic obesity 15. RM-718, described by Rhythm Pharmaceuticals as a "next-generation MC4R agonist" is a synthetic heptamer (seven amino-acid peptide) delivered subcutaneously weekly 15. Unlike setmelanotide, which can activate melanocortin receptors MC3R and MC1R in addition to MC4R 36, RM-718 by design, avoids stimulating MC1R 40 and thereby avoids the common hyperpigmentation adverse event 41 that can arise when MC1R is stimulated 42. RM-718 is currently in Phase I for obesity and hypothalamic obesity indications, 43.

As an alternative to injectables, Rhythm Pharmaceuticals is also developing bivamelagon, (LB54640), for daily oral delivery 44. LB54640 is a small molecule MC4R agonist licenced from LG Chem Ltd. 15 presently in Phase II clinical trials for hypothalamic obesity 45. Preclinical and Phase I work suggests that LB54640 should not cause hyperpigmentation 44.

Palatin Technologies is targeting selective agonism of melanocortin receptors to treat inflammatory and autoimmune conditions, with a particular focus on ocular indications 46. In 2019, the company received FDA-approval for Vyleesi (bremelanotide acetate), an injectable α-MSH analogue, making it the first FDA-approved melanocortin agent 15, 47. While Vyleesi is indicated for Hypoactive Sexual Desire Disorder (HSDD) in premenopausal women 15, bremelanotide is also in development as an obesity treatment. Topline results from a recent Phase II study 48 in which bremelanotide was co-administered alongside tirzepatide suggest that bremelanotide may help to prevent weight regain, a known issue after tirzepatide treatment ceases 49, 50. Palatin Technologies has two additional MC4R agonists, PL-7737 which is an oral small molecule, and PL-8905 which is a synthetic peptide, both in preclinical development for obesity indications, with the former specifically targeting obesity caused by LEPR deficiency 15.

↑ Back to top

6. Other Anti-Obesity Drugs

Qsymia was approved in 2012 15 and includes two active agents, phentermine and topiramate, in an extended-release capsule 51. Qsymia is used in combination with recommendations for diet and physical activity to achieve weight loss in obese individuals at least 12 years old and in overweight adults with at least one weight-related comorbidity. One of the active ingredients, phentermine, inhibits reuptake of serotonin, noradrenaline and dopamine, effectively increasing their concentration and leading to appetite suppression or feelings of satiety 52, 53. The other active agent in Qsymia, topiramate, is a neurostabiliser that achieves weight loss via an unknown mechanism of action 54, 55. During its use in seizures and migraine, topiramate-induced weight-loss was considered a major adverse event 15, 55. Contributing factors for weight-loss may include dysgeusia 56 and GABA activation culminating in appetite suppression and increases in satiety 57.

Like Qsymia, Contrave, (bupropion hydrochloride + naltrexone), marketed in the US since 2014, is also a combination of two active ingredients delivered orally in an extended-release tablet 15. The drug is an anorexiant designed to reduce hunger and cravings 58, and is used as a medical support to diet and exercise interventions for weight loss in overweight individuals with a weight-related co-morbidity or obesity 58. Its active ingredients are small molecules that have been individually marketed as drugs for applications other than weight loss 58. The precise mechanism of action of Contrave is unknown though it is believed to work by targeting the hypothalamic melanocortin system (Figure 2) and the mesolimbic reward system 59. Bupropion, a dopamine reuptake inhibitor 15, activates POMC neurons to secrete neurotransmitters that reduce the desire to eat, and naltrexone, an opioid antagonist 15, blocks the autoinhibitory feedback loop in the hypothalamus to keep POMC neurons activated 58, 59.

↑ Back to top

7. Sales, Market Size and Opportunity

The US obesity market is buoyant and has not yet peaked. The US market alone, which was worth ~$12B in 2024 is expected to be worth ~ $149B by 2031 15.

Global annual sales for Zepbound, which were ~$4.9B USD in 2024 are forecast to grow to ~$26B-$28B USD by 2031, most of which is expected to be achieved through significant US sales (~$27.8B USD) versus the rest of the world, (~$204M USD).

Although Wegovy reported greater global annual sales than Zepbound in 2024, (~$8.4B USD), global annual sales for Wegovy (~$22B USD) are forecast to be similar but lower than Zepbound (~$26B-$28B USD) by 2031. This is likely due to competition from Zepbound in the US, where annual sales of Wegovy are projected to peak at ~$13B USD by 2027, which is approximately half of the anticipated peak sales value for Zepbound. The ex-US market for Wegovy is anticipated to generate ~$10B USD of annual sales by 2031.

Sales of setmelanotide are forecast to exceed $1B USD by 2030 15, with the US market expected to represent ~80% of global sales. Annual global sales of Qsymia and Contrave are comparatively modest. Nevertheless, Contrave has recently reported increasing its year-on-year revenues for two consecutive years, citing greater access in the US and expansion into other markets as the reasons behind its recent sales growth 60. A patient-based forecast of sales for Qsymia in obesity anticipates year-on-year increases in sales to the end of the forecast period (2031), when annual US sales are expected to reach $58M.

The market is set to expand as more treatment options are approved. For example, the availability of oral formulations and the approval of drugs with greater efficacy and durability are likely to be favoured by patients, and by offering alternative treatment options, new drugs will likely expand the overall patient population eligible for weight-loss pharmacotherapy.

↑ Back to top

8. Future Prospects

Though it is widely acknowledged that being overweight generally increases an individual's risk of ill-health, weight-loss is often difficult to achieve through willpower alone and pharmaceutical support to aid weight-loss has existed since the mid-twentieth century. In recent years, the emergence of safe and effective weight-loss pharmacotherapy has offered patients non-invasive alternatives to bariatric surgery to restore a healthy body weight and metabolic status.

Despite the evident success of marketed weight-loss pharmacotherapies like peptide drugs, developers realise that patients would prefer alternatives to regular dosing by subcutaneous injection, which is currently the most commonly available route of administration. Some of the most advanced drug candidates in the pipeline, NN-9932 (Novo Nordisk), cagrilintide (Novo Nordisk) and orforglipron calcium (Eli Lilly) are intended for oral delivery. Long-acting formulations that would increase the intervals between dosing would also be attractive to patients and offer a significant market advantage to drug developers.

Drugs with mechanisms of action that differ from or build upon those of already marketed drugs are also promising. For example, amylin receptor (AMYR) agonists are an emerging class of anti-obesity medications. The AMYRs are heterodimers formed by combining a cell surface expressed calcitonin G protein coupled receptor (CTR) with any one of three transmembrane proteins, known as receptor activity modifying proteins (RAMPs). Therefore, three different AMYR subtypes exist. Binding with a RAMP alters the structure and function of the CTR. Consequently, the AMYR has a great affinity for amylin, but not for calcitonin, (a hormone that regulates calcium and phosphate levels in blood).

Amylin is a 37 amino-acid polypeptide gut hormone that regulates satiety postprandially. It is co-secreted with insulin from the pancreas upon arrival of nutrients to the small intestine and regulates glucose homeostasis by slowing gastric emptying, suppressing glucagon secretion and ultimately suppressing hunger. In the hypothalamus, amylin can work synergistically with, or independent of leptin to affect anorexigenic signalling.

Late-stage drug candidates that agonise AMYRs include cagrilintide, and the combination drug CagiSema, (Novo Nordisk). Others in earlier clinical development are AZD-6234 (AstraZeneca), eloralintide (Eli Lilly), petrelintide (Eli Lilly via Zealand Pharma) and NN-9490 (Novo Nordisk).

Drug products formed by combining anti-obesity agents is also promising. This approach allows for repurposing of approved drugs and may improve potency by invoking multiple mechanisms of action. The most advanced of these, CagriSema (Novo Nordisk) is at Phase III and combines cagrilintide, a lipidated amylin analogue with semaglutide. At Phase I, (insulin lispro + pramlintide) (Adocia SAS) combines a fast-acting insulin formulation with pramlintide, an amylinomimetic drug marketed for T2D, while the combination of two ultra long-acting peptides, MET-233i, a nutrient-stimulated hormone analogue that targets amylin, with MET-097i, a GLP1RA, form another drug product, (MET-233i + MET-097i) (Metsera Inc).

One of the potentially undesirable side-effects of weight-loss drugs is loss of lean mass, which, if excessive, could increase an individual's frailty and reduce their strength and functionality. Lean mass loss was reported for patients on semaglutide (~10% loss relative to baseline) and tirzepatide (~11% loss relative to baseline) during Phase III clinical trials, STEP-1, NCT03548935 and SURMOUNT-1, NCT04184622, respectively 61, 62. Notably, in SURMOUNT-1, lean mass loss as a proportion of total mass lost was similar (~25%) across the treatment and placebo groups62, and in the treatment group, physical function scores were improved relative to baseline and by comparison to the placebo group at the end of the study 62, 63. This suggested that the lean mass loss was generally not debilitating. Moreover, despite the lean-mass losses, STEP-1 reported an overall improvement in the lean body mass:fat mass ratio, which may be indicative of improved physical condition.

Lending further support, a study of anti-diabetic drugs and sarcopenia risk found no significant association between GLP1RAs and sarcopenia (low skeletal muscle mass and poor muscle function) 64. Nevertheless, the question of whether loss of lean body mass is an issue of concern remains open 65, 66, 67.

Candidate drugs are in development for treatment of sarcopenic obesity, often a disease of aging and defined by the coexistence of obesity and sarcopenia 68. For example, Immunis Inc is developing IMM01-STEM for sarcopenic obesity 69 and Biophytis SA has progressed BIO101 (20-hydroxyecdysone) through a Phase I clinical trial 70 for age-related sarcopenia, including sarcopenic obesity.

Long-term durability of weight-loss from pharmacotherapy has yet to be mastered. Amongst a cohort of overweight or obese individuals with preexisting cardiovascular disease, weight-loss on semaglutide was sustainable for at least four years 71. In overweight and obese adults, weekly tirzepatide treatment yielded weight loss sustained over three years 72. Without lifestyle adjustments, however, rapid and substantial weight regain has been reported when these drug interventions were withdrawn 50, 73 reflective of the chronicity of obesity. Some options have successfully prevented weight regain in these scenarios, such as low dose bremelanotide (Palatin Technologies) used as a maintenance therapy following withdrawal of tirzepatide 48, 74.

Looking to the future, the availability of safe and effective weight loss pharmacotherapy will directly impact the health and wellbeing of obese individuals and has the potential to impact the delivery of population-level healthcare and influence public health messaging around weight-management. While good progress has already been made in bringing safe and efficacious weight-loss drugs to market, the field continues to evolve. Formulations compatible with oral dosing are expected to increase use of these medicines and to benefit patient compliance. Opportunities exist to develop drugs with a durable weight-loss effect in the absence of ongoing pharmacotherapy which would be greatly attractive to patients. Co-therapies and drugs that preserve lean-mass, prevent excessive lean-mass loss or that promote loss of fat- over lean- mass may also find a sizeable market. As we come to a more comprehensive understanding of the signalling and molecular mechanisms underlying obesity it is very likely that new drugs with novel mechanisms of action will also enter clinical development.

Overall, the medicine cabinet for treating obesity is filling up, demonstrating what scientific innovation and endeavour can deliver for complex and multi-factorial metabolic diseases.

 

↑ Back to top

9. References

  1. World Health Organization technical report series, 894, 2000, "Obesity: preventing and managing the global epidemic". ISBN 92 4 120894 5. https://iris.who.int/handle/10665/42330 252 p. 252 p.
  2. World Health Organization, 2023, "Health service delivery framework for prevention and management of obesity". ISBN 978-92-4-007323-4.
  3. Phelps et al., 2024, "Worldwide trends in underweight and obesity from 1990 to 2022: a pooled analysis of 3663 population representative studies with 222 million children, adolescents, and adults". Lancet, 403(10431):1027-1050. doi: 10.1016/S0140-6736(23)02750-2.
  4. Lobstein et al., 2022, "World Obesity Atlas 2022". https://www.worldobesity.org/resources/resource-library/world-obesity-atlas-2022
  5. Rubino et al., 2025, "Definition and diagnostic criteria of clinical obesity". Lancet Diabetes Endocrinol. 13(3)221-262. doi: 10.1016/S2213-8587(24)00316-4.
  6. Coleman 2005, "Anorectics on trial: a half century of federal regulation of prescription appetite suppressants". Ann Intern Med 143(5):380-5. doi: 10.7326/0003-4819-143-5-200509060-00013.
  7. Gadde and Atkins, 2020, "The limits and challenges of antiobesity pharmacotherapy". Expert Opin Pharmacother. 21(11):1319-1328. doi: 10.1080/14656566.2020.1748599.
  8. Haslam, 2016, "Weight management in obesity – past and present". Int J Clin Pract 70(3):206-17. doi: 10.1111/ijcp.12771.
  9. James et al., 2010, "Effect of sibutramine on cardiovascular outcomes in overweight and obese subjects". N Engl J Med 363(10):905-17. doi: 10.1056/NEJMoa1003114.
  10. Onakpoya et al., 2016, "Post-marketing withdrawal of anti-obesity medicinal products because of adverse drug reactions: a systematic review". BMC Med 14(1):191. doi: 10.1186/s12916-016-0735-y.
  11. James et al., 2005, "The SCOUT study: risk-benefit profile of sibutramine in overweight high-risk cardiovascular patients". Eur Heart J Suppl. (7)L44-L48. doi.org/10.1093/eurheartj/sui086
  12. Gustafson et al., 2013, "Lorcaserin (Belviq) A selective serotonin 5-HT2C agonist in the treatment of obesity", PT 38(9):525-34. PMID: 24273398
  13. S. Food and Drug Administration, (2020), "FDA requests the withdrawal of the weight-loss drug Belviq, Belviq XR (lorcaserin) from the market". https://www.fda.gov/drugs/drug-safety-and-availability/fda-requests-withdrawal-weight-loss-drug-belviq-belviq-xr-lorcaserin-market. Accessed 19th May 2025.
  14. Sharma et al., 2006, "The Etiology of Hypertension in the Metabolic Syndrome Part Two: The Gene-Environment Interaction". Curr Vasc Pharmacol 4(4):305-20. doi: 10.2174/157016106778521661.
  15. GlobalData, https://www.globaldata.com/
  16. Nauck et al., 2021, "The evolving story of incretins (GIP and GLP-1) in metabolic and cardiovascular disease: A pathophysiological update". Diabetes Obes Metab 23:Suppl 3:5-29 doi: 10.1111/dom.14496.
  17. Sallam et al., 2024, "Shifting the Scales: Tirzepatide's breakthrough in obesity management". Cureus 16(5):e60545. doi: 10.7759/cureus.60545.
  18. Deacon and Ahrén, 2011, "Physiology of incretins in health and disease". Rev Diabet Stud 8(3):293-306. doi: 10.1900/RDS.2011.8.293.
  19. Zheng et al., 2024, "Glucagon-like peptide-1 receptor: mechanisms and advances in therapy". Signal Transduct Target Ther 991):234. doi: 10.1038/s41392-024-01931-z.
  20. Knudsen and Lau, 2019, "The discovery and development of liraglutide and semaglutide". Front Endocrinol (Lausanne). 12:10:155. doi: 10.3389/fendo.2019.00155. eCollection 2019.
  21. Frequently Asked Questions about Wegovy, https://www.wegovy.com/tools-and-resources/faq.html. Accessed 19th May 2025.
  22. Sun et al., 2022, "Structural determinants of dual incretin receptor agonism by Tirzepatide". Proc Natl Acad Sci USA 119(13)e2116506119. doi: 10.1073/pnas.2116506119.
  23. Aronne et al., 2025, "Tirzepatide as compared with semaglutide for the treatment of obesity". N Engl J Med, Online ahead of print, doi: 10.1056/NEJMoa2416394.
  24. Amin et al., 2024, "Evaluation of an oral small-molecule glucagon-like peptide-1 receptor agonist, lotiglipron, for type 2 diabetes and obesity: A dose-ranging, phase 2, randomized, placebo-controlled study". Diabetes Obes Metab. 27(1):215-227. doi: 10.1111/dom.16005.
  25. Pfizer, 2025, "Pfizer provides update on oral GLP-1 receptor agonist Danuglipron". https://www.pfizer.com/news/press-release/press-release-detail/pfizer-provides-update-oral-glp-1-receptor-agonist. Accessed 19th May 2025.
  26. Obesity and overweight, https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight. Accessed 4th June 2025.
  27. Causes Obesity, https://www.nhs.uk/conditions/obesity/causes/. Accessed 4th June 2025.
  28. Gong, 2014, "Leveraging melanocortin pathways to treat glomerular diseases", Adv Chronic Kidney Dis 21(2):134-51. doi: 10.1053/j.ackd.2013.09.004.
  29. Yeo et al., 2021, "The melanocortin pathway and energy homeostasis: From discovery to obesity therapy". Mol Metab 48:101206. doi: 10.1016/j.molmet.2021.101206.
  30. Harno et al., 2018, "POMC: The physiological power of hormone processing". Physiol Rev 98(4):2381-2430. doi: 10.1152/physrev.00024.2017.
  31. Takahashi and Mizusawa, 2013, "Posttranslational modifications of proopiomelanocortin in vertebrates and their biological significance". Front Endocrinol (Lausanne). 17(4):143 doi: 10.3389/fendo.2013.00143.
  32. Wabitsch et al., 2022, "Natural history of obesity due to POMC, PCSK1, and LEPR deficiency and the impact of setmelanotide". J Endocr Soc 6(6)bvac057. doi: 10.1210/jendso/bvac057.
  33. Imcivree mechanism of action, https://www.imcivree.com/hcp/ppl/moa/. Accessed 19th May 2025.
  34. S. Food and Drug Administration, (2022), "FDA approves treatment for weight management in patients with Bardet-Biedl Syndrome aged 6 or older". https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-treatment-weight-management-patients-bardet-biedl-syndrome-aged-6-or-older#:~:text=The%20U.S.%20Food%20and%20Drug,%2DBiedl%20Syndrome%20(BBS). Accessed 19th May 2025.
  35. Rhythm Pharmaceuticals, 2024, "Rhythm Pharmaceuticals announces FDA approval of IMCIVRE® (setmelanotide) for patients as young as 2 years old". https://ir.rhythmtx.com/news-releases/news-release-details/rhythm-pharmaceuticals-announces-fda-approval-imcivreer-0. Accessed 19th May 2025.
  36. Hussain and Farzam, 2023, "Setmelanotide". PMID: 36943959.
  37. IUPHAR/BPS guide to pharmacology, "Setmelanotide", https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=structure&ligandId=9272#. Accessed 19th May 2025.
  38. Collet et al., 2017 "Evaluation of a melanocortin-4 receptor (MC4R) agonist (Setmelanotide) in MC4R deficiency". Mol Metab 6(10):1321-1329. doi: 10.1016/j.molmet.2017.06.015.
  39. Qamar et al., 2024, "Setmelanotide: A melanocortin-4 receptor agonist for the treatment of severe obesity due to hypothalamic dysfunction". touchREV Endocrinol 20(2):62-71. doi: 10.17925/EE.2024.20.2.9.
  40. Rhythm Pharmaceuticals, 2024, "Rhythm Pharmaceuticals announces first patient dosed in Phase 1 trial evaluating RM-718, a weekly MC4R-specific agonist". https://www.globenewswire.com/news-release/2024/03/25/2851546/0/en/Rhythm-Pharmaceuticals-Announces-First-Patient-Dosed-in-Phase-1-Trial-Evaluating-RM-718-a-Weekly-MC4R-specific-Agonist.html. Accessed 19th May 2025.
  41. Haws et al., 2020 "Effect of setmelanotide, a melanocortin-4 receptor agonist, on obesity in Bardet-Biedl syndrome". Diabetes Obes Metab. 22(11):2133-2140. doi: 10.1111/dom.14133.
  42. Bohm et al., 2025, "An overview of benefits and risks of chronic melanocortin-1 receptor activation". J Eur Acad Dermatol Venereol. 39(1):39-51. doi: 10.1111/jdv.20269.
  43. gov, "A Study of RM-718 in healthy subjects and in patients with HO". NCT06239116.
  44. Rhythm Pharmaceuticals, 2024, "Rhythm Pharmaceuticals announces first patients dosed in Phase 2 Trial evaluating oral MC4R agonist LB54640 in hypothalamic obesity." https://www.globenewswire.com/news-release/2024/07/23/2917160/0/en/Rhythm-Pharmaceuticals-Announces-First-Patients-Dosed-in-Phase-2-Trial-Evaluating-Oral-MC4R-Agonist-LB54640-in-Hypothalamic-Obesity.html. Accessed 19th May 2025.
  45. gov "A Study to Assess Efficacy and Safety of LB54640 in Patients With Hypothalamic Obesity", NCT06046443.
  46. Palatin Technologies, "Our science", https://palatin.com/our-science/. Accessed 19th May 2025.
  47. Palatin Technologies, "Our pipeline", https://palatin.com/pipeline/. Accessed 19th May 2025.
  48. gov, "A Phase 2 study evaluating the co-administration of bremelanotide with tirzepatide for the treatment of obesity", NCT06565611.
  49. Palatin Technologies, 2025, "Palatin reports positive appetite suppression results from Phase 2 obesity study of MC4R agonist bremelanotide and tirzepatide". https://palatin.com/press_releases/palatin-reports-positive-appetite-suppression-results-from-phase-2-obesity-study-of-mc4r-agonist-bremelanotide-and-tirzepatide/. Accessed 19th May 2025.
  50. Aronne et al., 2024, "Continued treatment with tirzepatide for maintenance of weight reduction in adults with obesity: The SURMOUNT-4 randomized clinical trial". JAMA 331(1):38-48. doi: 10.1001/jama.2023.24945.
  51. com, "Qsymia", https://www.drugs.com/qsymia.html. Accessed 20th May 2025.
  52. com, "Phentermine Patient Tips", https://www.drugs.com/tips/phentermine-patient-tips#. Accessed 20th May 2025.
  53. Inxight Drugs, "Phentermine", https://drugs.ncats.io/drug/C045TQL4WP#. Accessed 20th May 2025.
  54. Shin and Gadde, 2013, "Clinical utility of phentermine/topiramate (Qsymia™) combination for the treatment of obesity". Diabetes Metab Syndr Obes 8(6)131-9. doi: 10.2147/DMSO.S43403.
  55. Wajid et al., 2023, "Topiramate (Topamax): evolving role in weight reduction management: A narrative review". Life (Basel), 13(9)1845. doi: 10.3390/life13091845.
  56. Vivus LLC, 2024, "Highlights of prescribing information" PH-03-002-16, Revised: 09/2024. https://qsymia.com/patient/include/media/pdf/prescribing-information.pdf?v=250220.
  57. How Qsymia works, https://hcp.qsymia.com/how-qsymia-works/. Accessed 20th May 2025.
  58. How Contrave works. https://contrave.com/how-contrave-works/. Accessed 4th June 2025.
  59. Ornellas et al., 2011, "Naltrexone SR/Bupropion SR (Contrave): A new approach to weight loss in obese adults". P T 36(5):255-62. PMID: 21785538
  60. Currax announces outstanding 2024 financial and operational performance. curraxpharma.com. Accessed 23rd June 2025.
  61. Wilding et al., 2021, "Impact of semaglutide on body composition in adults with overweight or obesity: exploratory analysis of the STEP 1 study". J Encdocrin Soc A16. doi: 10.1210/jendso/bvab048.
  62. Look et al., 2025, "Body composition changes during weight reduction with tirzepatide in the SURMOUNT-1 study of adults with obesity or overweight". Diabetes Obes Metab 27(5):2720-2729. doi: 10.1111/dom.16275.
  63. Gudzune et al., 2025 "Association between weight reduction achieved with tirzepatide and quality of life in adults with obesity: Results from the SURMOUNT-1 study". Diabetes Obes Metab. 27(2):539-550.  doi: 10.1111/dom.16046.
  64. Kong et al., 2025, "Worldwide burden of antidiabetic drug-induced sarcopenia: An international pharmacovigilance study". 129:105656. doi: 10.1016/j.archger.2024.105656.
  65. Prado et al., 2024, "Muscle matters: the effects of medically induced weight loss on skeletal muscle". Lancet Diabetes Endocrinol. 12(11):785-787. doi: 10.1016/S2213-8587(24)00272-9.
  66. Hope et al., 2024, "Skeletal muscle loss and sarcopenia in obesity pharmacotherapy". 20(12):695-696. doi: 10.1038/s41574-024-01041-4.
  67. Neeland et al., 2024, "Changes in lean body mass with glucagon-like peptide-1-based therapies and mitigation strategies". Diabetes Obes Metab. Suppl 4:16-27. doi: 10.1111/dom.15728.
  68. Donini et al., 2022, "Definition and diagnostic criteria for sarcopenic obesity: ESPEN and EASO consensus statement". Obes Facts 15(3):321-335.
  69. gov, "Safety and efficacy of IMM01-STEM against placebo on muscle performance in seniors with obesity and muscle weakness". NCT06600581.
  70. gov, "A double-blind, placebo-controlled, randomized INTerventional Clinical Trial (SARA-INT) (SARA-INT)". NCT03452488.
  71. Ryan et al., 2024, "Long-term weight loss effects of semaglutide in obesity without diabetes in the SELECT trial". Nat Med. 30(7):2049-2057.
  72. EurekAlert!, 2025, "Tirzepatide can produce clinically meaningful weight loss for at least 3 years in adults with overweight or obesity who don't have diabetes". https://www.eurekalert.org/news-releases/1080117. Accessed 20th May 2025.
  73. Wilding et al., 2022, "Weight regain and cardiometabolic effects after withdrawal of semaglutide: The STEP 1 trial extension". Diabetes Obes Metab. 24(8):1553-1564.
  74. PR Newswire 2025, "Palatin reports positive appetite suppression results from Phase 2 obesity study of MC4R agonist bremelanotide and tirzepatide".https://www.prnewswire.com/news-releases/palatin-reports-positive-appetite-suppression-results-from-phase-2-obesity-study-of-mc4r-agonist-bremelanotide-and-tirzepatide-302431194.html. Accessed 20th May 2025.

↑ Back to top