Skip to main content

OPINION article

Front. Endocrinol., 05 June 2024
Sec. Obesity
This article is part of the Research Topic Insights in Obesity: 2023 View all 8 articles

Semaglutide for weight loss: unanswered questions

Ploutarchos Tzoulis*Ploutarchos Tzoulis1*Stephanie E. BaldewegStephanie E. Baldeweg2
  • 1Department of Metabolism & Experimental Therapeutics, Division of Medicine, University College London, London, United Kingdom
  • 2Division of Medicine, University College London, London, United Kingdom

Introduction

Obesity, a chronic disease associated with significant morbidity (1)and excess mortality (2), has become a global epidemic with a significant impact on healthcare systems and society in general (2). Lifestyle interventions, the cornerstone of treatment for obesity, have limited long-term efficacy, with maintenance of weight loss being the main challenge (35). The history of anti-obesity medications (AOMs) has been marked by withdrawal of more than 20 medications for safety reasons (6). Amongst the earliest AOMs developed, amphetamine derivatives were used widely since 1950s, but most, except for phentermine, have subsequently been withdrawn due to their cardiovascular risk and abuse potential (7). In recent years, the most prominent examples of AOMs failures have been sibutramine, rimonabant, and lorcaserin which were withdrawn secondary to adverse cardiovascular events, psychiatric safety, and increased occurrence of cancer, respectively (6, 8). Finally, the example of benfluorex, a medication used off-label for weight loss which, despite several case reports of cardiac valvulopathy, was withdrawn after significant delay (9), has highlighted the importance of pharmacovigilance. Until recently, AOMs available in Europe; namely, orlistat, combined naltrexone and bupropion, and liraglutide, a daily injectable glucagon-like peptide 1 (GLP-1) agonist (1); were underutilised in clinical practice (10, 11)since they produced a mean placebo-subtracted weight loss of around 5% (12), lower than the weight loss required to ameliorate most weight-related compications (12, 13). Semaglutide, a weekly injectable GLP-1 agonist widely used for the treatment of type 2 diabetes mellitus (DM), has been approved since 2021 by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for chronic weight management in patients with obesity or body mass index (BMI) above 27 kg/m2andat least one weight-related condition. Its weight-lowering effect is based on appetite reduction and satiety increase through activation of GLP-1 receptors in the hypothalamus and the hindbrain (14, 15). In comparison to other available AOMs, semaglutide shows a considerable enhancement in efficacy, being the first medication safely producing a greater than 10% average weight loss over that attributable to lifestyle interventions (12). A comprehensive programme of phase 3 randomised clinical trials (RCTs), the Semaglutide Treatment Effect in People with Obesity (STEP) trials, has evaluated the effect of semaglutide use for weight management in people with overweight or obesity, reporting a mean placebo-subtracted percentage weight loss of 12.5%, with more than half of participants achieving weight reduction of 15% or more from baseline (12). The unprecedented efficacy of semaglutide for weight loss has generated widespread publicity and led to global drug shortages. However, numerous research questions have arisen in relationship to its optimal use.

What is the optimal duration of semaglutide therapy for weight loss?

The longest available efficacy data for semaglutide as an AOM are derived from the STEP 5 trial, showing a durable, sustained, substantial weight loss over a 2-year study duration with mean reduction in body weight of 15.2% in semaglutide group compared to 2.6% in placebo group (16). Both reduction in weight and improvement in cardiometabolic parameters, such as HbA1c, blood pressure and lipid profile, reached a plateau after 60 weeks and were sustained with its continued use for another year (16). Bearing in mind that obesity is a chronic, progressive, relapsing disease (17), the optimal duration of semaglutide therapy for weight management remains to be determined and may need to be tailored to individual characteristics. Future studies are required to investigate the durability of weight loss and metabolic benefits over different lengths of therapy and in different subgroups.

Is weight regain after semaglutide discontinuation predictable and preventable?

In general, any successful strategy to tackle the obesity epidemic should start with the recognition that the modern obesogenic environment plays a key role, necessitating a societal approach (18). Implementation of public health policies is essential, for example lowering the content of trans fatty acids in food, promoting healthier diets and regular physical exercise in the school setting, and providing subsidies for healthy food choices combined with imposing taxation on sugar-sweetened beverages (18). Increasingly popular ‘‘cafeteria’’ or ‘‘junk food’’ diets, consisting of ultra-processed foods which contain high levels of refined sugar and saturated fat, lead to hyperphagia and obesity (19), highlighting the need for initiatives, such as user-friendly nutritional labelling of food products in order to empower people with the right information to make healthier choices. At a personal level and with respect to pharmacotherapy, significant weight gain after semaglutide discontinuation has been reported in two studies, STEP 4 (20) and STEP 1 trial extension (21). Individuals who discontinued semaglutide after 20 and 68 weeks regained over the next year off-pharmacotherapy 50% and 63% of the prior weight loss, respectively (20, 21). It is unclear whether this trajectory of weight regain continues in the subsequent years and to which extent, if any, a final net weight loss is achieved. More rapid and greater weight regain was observed in individuals who had achieved greater weight loss (21) and also when lifestyle intervention was withdrawn, such as in the STEP 1 trial extension (21), compared with the continuing provision of lifestyle intervention, such as in STEP 4 (20). These findings highlight the need for real-life withdrawal studies to identify the predictors of the rapidity and magnitude of weight gain and investigate the effect of continuing counselling and lifestyle modification on the trajectory of weight regain (21). Different strategies to prevent or, at least, ameliorate this phenomenon should be explored, including down titration of semaglutide dose, enrolling people in specific lifestyle programmes after drug discontinuation, and adopting specific criteria for another treatment course of semaglutide in case of rapid weight regain.

Can we predict response to semaglutide?

All STEP trials have reported a marked variability in semaglutide response with 32–39.6% of people being ‘‘super responders’’, achieving weight loss in excess of 20%, and a subgroup of 10.2–16.7% of individuals being ‘‘non-responders’’, reducing body weight by less than 5% from baseline (16, 20, 22, 23).

Two predictors of lower weight loss with semaglutide have been identified; the coexistence of type 2 DM, with a mean body weight decrease of 14.9% in those without DM (22) versus 9.6% in those with DM (24), and the male gender, with an average 8–9.3% weight loss in males compared to 14–16.2% in females (25). The great heterogeneity in semaglutide-related weight loss highlights the need for studies examining the predictive role of demographic characteristics (gender, ethnic origin, age), metabolic parameters (baseline BMI, glycated haemoglobin, fasting glucose, markers of insulin resistance, lipid profile), eating behaviours (levels of hunger, satiety, episodes of hyperphagia, cravings) and genotype for weight response to semaglutide. Emerging evidence suggests that phenotype-guided, pathophysiology-based use of AOMs may enhance weight loss outcomes of pharmacotherapy, recommending GLP-1 agonists as the treatment of choice for people with ‘‘hungry gut’’ phenotype who exhibit abnormal satiety with reduced duration of fullness and rapid gastric emptying (26). Precision medicine initiatives, integrating data from genetics, epigenetics and metabolomics as well as encompassing demographic, environmental and psychological factors, are currently in progress in order to stratify obesity into different phenotypes with distinct underlying physiology and different risk for future complications (27, 28). Future studies will determine whether personalised decision-making about obesity pharmacotherapy, based on predictive models and novel algorithms, could optimise therapeutic benefit and minimise risks (27, 29). The advent of several novel AOMs, expected to be approved for clinical use in the near future, suggests a key role for prediction tools in day-to-day care to identify non-responders to semaglutide who would benefit more from alternative pharmacotherapies, such as dual GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) agonist (30) or combination of GLP-1 agonist an amylin analogue (31) or triple GLP-1, GIP and glucagon receptors agonist (32), or endoscopic procedures or bariatric surgery (27).

Which BMI cut-off should be used to consider semaglutide prescription for weight loss?

BMI, widely adopted as the marker used to define overweight and obesity, remains the main eligibility criterion for semaglutide administration. However, BMI is not an accurate measure of adipose tissue in the body and has two major limitations to diagnose obesity in an individual (33); firstly, it cannot distinguish fat from lean body mass which requires the use of dual-energy X-ray absorptiometry or bioelectric impedance; second, it cannot provide information about body fat distribution which would be feasible with use of anthropometric indices, such as waist circumference (17). Semaglutide administration for weight management has been approved for BMI greater than 30 kg/m2, regardless of comorbidities, or a BMI above 27 kg/m2 with at least one weight-related complication, producing similar percentage weight loss across different categories of baseline BMI (34). Taking into account the high prevalence of obesity, the high cost of semaglutide and its substantial budget impact, state and private health insurance providers will use varying BMI cut-offs as the criterion for reimbursement. For example, recent guidance in the United Kingdom recommends semaglutide as an option for weight management in people with BMI of at least 35 kg/m2, or alternatively a BMI of at least 30 kg/m2 with at least one weight-related complication. Therefore, cost-effectiveness analysis is essential in order to determine the optimal BMI range, across other criteria, which would ensure the most appropriate use of limited healthcare resources. Future studies need to evaluate whether predictive models, incorporating various parameters, should replace BMI as predictors of those at the greatest risk of complications who would benefit most from weight loss (27).

What is the optimal lifestyle intervention as adjunct to semaglutide use?

In general, all people with obesity should receive individualised medical nutrition therapy and engage in regular physical activity (35). Any of multiple nutrition interventions can be considered with personalisation and long-term adherence being essential for sustained weight loss (35). With regards to the weight loss outcomes in patients treated with semaglutide as an adjunct to standard lifestyle intervention (16, 20, 22, 36) they were similar with those observed in STEP 3 trial in combination with intensive lifestyle modification (23), questioning the additional benefit of the inclusion of an initial 8-week meal-replacement phase or intensive behavioural therapy (23). Two trials, starting with an initial 12-week intensive lifestyle intervention followed by liraglutide (37) or tirzepatide, a dual GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) agonist (38), produced additive weight loss which approached the combined results of each intervention on its own (37, 38),, suggesting that sequential use may be superior to concurrent use (38). The semaglutide-induced physiologically driven reduction in calorie intake puts the scope and intensity of lifestyle intervention into question (38), highlighting the need for prospective head-to-head studies comparing sequential with concurrent use of semaglutide across intensive caloric restriction diet. Finally, prospective studies need to compare the weight loss outcomes of semaglutide in combination with different diet regimens, varying on macronutrient composition and meal frequency, for example time-restricted eating.

What is the role of semaglutide before and after bariatric surgery?

Weight regain following bariatric surgery is common (39), with numerous studies showing that liraglutide leads to significant weight reduction in these individuals (4043). Due to their exclusion from the STEP programme, there is lack of high-quality evidence about semaglutide use in post-bariatric surgery populations. However, three retrospective real-world studies support the effectiveness and good safety profile of semaglutide for the treatment of weight recurrence after bariatric surgery, as evidenced by a mean 6-month body weight reduction of 9.8–10.3% (4446). Ongoing prospective studies, such as the BARI-STEP trial, aim to determine the role of semaglutide in individuals who have experienced insufficient weight loss or excessive weight regain following bariatric surgery (47, 48). Another potential, albeit untested yet, therapeutic application of semaglutide is that of neoadjuvant pharmacotherapy prior to bariatric surgery. Prospective studies are warranted to determine whether semaglutide use in the preoperative setting could improve patient outcomes and reduce complications of bariatric operations, especially in individuals with very high BMI exceeding 50 kg/m2. Finally, future research efforts should also be directed towards the combination of semaglutide with, rapidly evolving, endoscopic bariatric therapies, such as intragastric balloons or endoscopic sleeve gastrectomy (48).

What is the efficacy and safety of semaglutide for weight management in patients with active psychiatric disease?

Psychiatric conditions and obesity frequently occur together and have a bidirectional relationship (49), highlighting the need to generate specific data about semaglutide use in this context. Mental illness and obesity share not only pathogenic pathways involving the immune and endocrine system, but also coping behaviours and sociodemographic factors (50). Finally, it is worth mentioning that peptidergic systems critical to the regulation of energy homeostasis are also involved in neurocognition, as it applies to neuropeptide Y (NPY) and melanin-concentrating hormone (MCH) whose manipulation affects both appetite and diverse range of cognitive functions (51). Several psychotropic medications induce weight gain, contributing to high prevalence of obesity in people with psychiatric diseases (52). In individuals treated with antidepressants, semaglutide seems to retain its efficacy, as suggested by a post hoc exploratory analysis of STEP trials reporting a semaglutide-induced clinically meaningful weight loss, regardless of baseline antidepressant use (52). Antipsychotic-induced weight gain (AIWG), a common problem leading to chronic complications, may respond favourably to semaglutide, based on a small case series of patients with metformin-refractory AAWG (53). The lack of effective treatment options highlights the need for RCTs evaluating the effect of semaglutide on weight and metabolic parameters in patients with iatrogenic weight gain, such as olanzapine- and clozapine-treated patients with schizophrenia (54). Since STEP trials excluded patients who had major depressive disorder or bipolar disorder or schizophrenia within last two years prior to study enrollment (52), prospective studies are urgently needed to evaluate semaglutide use in patients with active mental health illness. Besides the need for efficacy outcomes in this population, the neuropsychiatric safety of semaglutide warrants evaluation in light of postmarketing reports about suicidal thoughts and behaviours in patients receiving semaglutide. Recent findings from a retrospective cohort study of patients receiving anti-obesity pharmacotherapy, reporting that semaglutide was associated with a significantly lower risk for suicidal ideation compared to alternative, non-GLP-1 agonists, AOMs (55), provide some reassurance. However, prospective studies are warranted to confirm the neuropsychiatric safety of semaglutide.

What is the long-term safety profile of semaglutide?

Semaglutide-induced gastrointestinal events, albeit common, are transient, mild to moderate in severity, include gallbladder-related events, primarily cholelithiasis, and lead to drug discontinuation in 5.9–7.7% of patients (22, 23, 20, 34, 36, 56). In line with high-grade evidence supporting the absence of a link of GLP-1 agonists with pancreatitis, pancreatic cancer, medullary thyroid cancer, or other malignant neoplasms (5759), STEP trials have reported extremely low rates for acute pancreatitis, no cases of pancreatic cancer, and rates of malignant neoplasms similar with those in placebo group (16, 20, 22, 23, 36). Also, a meta-analysis of all semaglutide studies has excluded an association of semaglutide with an increased risk of any types of cancer (60). Contrary to these reassuring safety data, a cohort study of people receiving pharmacotherapy for weight management indicated a significantly increased risk for pancreatitis, bowel obstruction and gastroparesis in those treated with GLP-1 agonists (61), including a 5-fold and 9-fold increased likelihood to develop pancreatitis and gastroparesis in semaglutide-treated patients, respectively (61). Additionally, postmarketing reports of ileus in patients on semaglutide have led the FDA to add a warning about gastrointestinal ileus on the semaglutide label, but a causal association has not been proven. In view of the very low background incidence and long latency period, clinical entities, such as ileus or pancreatic cancer, may go unnoticed in premarketing studies, necessitating post-marketing surveillance with various databases in order to draw definitive conclusions about a possible link with rare events (59). Finally, in line with any weight-lowering intervention (62, 63), semaglutide leads not only to reduction in fat mass, but also to reduction in total lean body mass and bone mineral density (22). These risks reinforce the importance of developing strategies to preserve muscle mass which, besides weight-bearing exercise and strength training, may entail novel pharmacological agents (63), namely bimagrumab, an activin antagonist (64), or myostatin inhibitors. Given the nature of obesity as a chronic, relapsing, progressive disease process, studies with long follow-up in combination with pharmacovigilance activities employing databases are warranted to ensure long-term safety of semaglutide and any other AOMs.

Conclusive remarks

In conclusion, the popularity of semaglutide has soared due to a combination of great efficacy and good safety profile. However, much remains to be learnt about the optimal use of semaglutide, highlighting the need for prospective studies, both controlled and real-world, exploring key unresolved issues, including its duration of treatment, predictors of response, appropriate type of lifestyle intervention, long-term safety profile and its use in specific settings. Semaglutide may herald a new era in the management of obesity, introducing widespread use of pharmacotherapy to meet a large unmet clinical need. Answering these questions will determine the extent of semaglutide use as part of the rapidly expanding armamentarium and facilitate personalised treatment of obesity.

Author contributions

PT: Conceptualization, Writing – original draft, Writing – review & editing. SB: Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Bray GA, Heisel WE, Afshin A, Jensen MD, Dietz WH, Long M, et al. The science of obesity management: an endocrine society scientific statement. Endocr Rev. (2018) 39:79–132. doi: 10.1210/er.2017-00253

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Chakhtoura M, Haber R, Ghezzawi M, Rhayem C, Tcheroyan R, Mantzoros CS. Pharmacotherapy of obesity: an update on the available medications and drugs under investigation. EClinicalMedicine. (2023) 58:101882. doi: 10.1016/j.eclinm.2023.101882

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Mann T, Tomiyama AJ, Westling E, Lew AM, Samuels B, Chatman J. Medicare’s search for effective obesity treatments: diets are not the answer. Am Psychol. (2007) 62:220–33. doi: 10.1037/0003-066X.62.3.220

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Dombrowski SU, Knittle K, Avenell A, Araujo-Soares V, Sniehotta FF. Long term maintenance of weight loss with non-surgical interventions in obese adults: systematic review and meta-analyses of randomised controlled trials. BMJ. (2014) 348:g2646

PubMed Abstract | Google Scholar

5. Enright C, Thomas E, Saxon DR. An updated approach to antiobesity pharmacotherapy: moving beyond the 5% Weight loss goal. J Endocr Soc. (2023) 7:bvac195

PubMed Abstract | Google Scholar

6. Onakpoya IJ, Heneghan CJ, Aronson JK. Post-marketing withdrawal of anti-obesity medicinal products because of adverse drug reactions: a systematic review. BMC Med. (2016) 14:191. doi: 10.1186/s12916-016-0735-y

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Rodgers RJ, Tschop MH, Wilding JP. Anti-obesity drugs: past, present and future. Dis Model Mech. (2012) 5:621–6. doi: 10.1242/dmm.009621

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Muller TD, Bluher M, Tschop MH, DiMarchi RD. Anti-obesity drug discovery: advances and challenges. Nat Rev Drug Discovery. (2022) 21:201–23. doi: 10.1038/s41573-021-00337-8

CrossRef Full Text | Google Scholar

9. Mullard A. Mediator scandal rocks French medical community. Lancet. (2011) 377:890–2. doi: 10.1016/S0140-6736(11)60334-6

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Saxon DR, Iwamoto SJ, Mettenbrink CJ, McCormick E, Arterburn D, Daley MF, et al. Antiobesity medication use in 2.2 million adults across eight large health care organizations: 2009–2015. Obes (Silver Spring). (2019) 27:1975–81. doi: 10.1002/oby.22581

CrossRef Full Text | Google Scholar

11. Claridy MD, Czepiel KS, Bajaj SS, Stanford FC. Treatment of obesity: pharmacotherapy trends of office-based visits in the United States from 2011 to 2016. Mayo Clin Proc. (2021) 96:2991–3000. doi: 10.1016/j.mayocp.2021.07.021

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Garvey WT. New horizons. A new paradigm for treating to target with second-generation obesity medications. J Clin Endocrinol Metab. (2022) 107:e1339–e47

PubMed Abstract | Google Scholar

13. Ryan DH, Yockey SR. Weight loss and improvement in comorbidity: differences at 5%, 10%, 15%, and over. Curr Obes Rep. (2017) 6:187–94

PubMed Abstract | Google Scholar

14. Blundell J, Finlayson G, Axelsen M, Flint A, Gibbons C, Kvist T, et al. 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) 19:1242–51.

PubMed Abstract | Google Scholar

15. 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) 23:754–62.

PubMed Abstract | Google Scholar

16. Garvey WT, Batterham RL, Bhatta M, Buscemi S, Christensen LN, Frias JP, et al. Two-year effects of semaglutide in adults with overweight or obesity: the STEP 5 trial. Nat Med. (2022) 28:2083–91. doi: 10.1038/s41591-022-02026-4

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Bray GA, Kim KK, Wilding JPH, World Obesity F. Obesity: a chronic relapsing progressive disease process. A position statement World Obes Federation. Obes Rev. (2017) 18:715–23. doi: 10.1111/obr.12551

CrossRef Full Text | Google Scholar

18. Temple NJ. A proposed strategy against obesity: how government policy can counter the obesogenic environment. Nutrients. (2023) 15(13):2910. doi: 10.3390/nu15132910

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Lalanza JF, Snoeren EMS. The cafeteria diet: A standardized protocol and its effects on behavior. Neurosci Biobehav Rev. (2021) 122:92–119. doi: 10.1016/j.neubiorev.2020.11.003

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Rubino D, Abrahamsson N, Davies M, Hesse D, Greenway FL, Jensen C, et al. Effect of continued weekly subcutaneous semaglutide vs placebo on weight loss maintenance in adults with overweight or obesity: the STEP 4 randomized clinical trial. JAMA. (2021) 325:1414–25

PubMed Abstract | Google Scholar

21. Wilding JPH, Batterham RL, Davies M, Van Gaal LF, Kandler K, Konakli K, et al. Weight regain and cardiometabolic effects after withdrawal of semaglutide: The STEP 1 trial extension. Diabetes Obes Metab. (2022) 24:1553–64. doi: 10.1111/dom.14725

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Wilding JPH, Batterham RL, Calanna S, Davies M, Van Gaal LF, Lingvay I, et al. Once-weekly semaglutide in adults with overweight or obesity. N Engl J Med. (2021) 384:989–1002. doi: 10.1056/NEJMoa2032183

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Wadden TA, Bailey TS, Billings LK, Davies M, Frias JP, Koroleva A, et al. Effect of subcutaneous semaglutide vs placebo as an adjunct to intensive behavioral therapy on body weight in adults with overweight or obesity: the STEP 3 randomized clinical trial. JAMA. (2021) 325:1403–13. doi: 10.1001/jama.2021.1831

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Davies M, Faerch L, Jeppesen OK, Pakseresht A, Pedersed SD, Perreault L, et al. Semaglutide 2.4 mg once a week in adults with overweight or obesity, and type 2 diabetes (STEP 2): a randomised, double-blind, double-dummy, placebo-controlled, phase 3 trial. Lancet. (2021) 397:971–84. doi: 10.1016/S0140-6736(21)00213-0

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Jensterle M, Rizzo M, Janez A. Semaglutide in obesity: unmet needs in men. Diabetes Ther. (2023) 14:461–5. doi: 10.1007/s13300-022-01360-7

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Acosta A, Camilleri M, Abu Dayyeh B, Calderon G, Gonzalez D, McRae A, et al. Selection of antiobesity medications based on phenotypes enhances weight loss: A pragmatic trial in an obesity clinic. Obes (Silver Spring). (2021) 29:662–71. doi: 10.1002/oby.23120

CrossRef Full Text | Google Scholar

27. Tahrani AA, Panova-Noeva M, Schloot NC, Hennige AM, Soderberg J, Nadglowski J, et al. Stratification of obesity phenotypes to optimize future therapy (SOPHIA). Expert Rev Gastroenterol Hepatol. (2023) 17(10):1031–9.

PubMed Abstract | Google Scholar

28. Abraham A, Yaghootkar H. Identifying obesity subtypes: A review of studies utilising clinical biomarkers and genetic data. Diabetes Med. (2023) 40:e15226. doi: 10.1111/dme.15226

CrossRef Full Text | Google Scholar

29. Bomberg EM, Ryder JR, Brundage RC, Straka RJ, Fox CK, Gross AC, et al. Precision medicine in adult and pediatric obesity: a clinical perspective. Ther Adv Endocrinol Metab. (2019) 10:2042018819863022. doi: 10.1177/2042018819863022

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Jastreboff AM, Aronne LJ, Ahmad NN, Wharton S, Connery L, Alves B, et al. Tirzepatide once weekly for the treatment of obesity. N Engl J Med. (2022) 387:205–16. doi: 10.1056/NEJMoa2206038

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Enebo LB, Berthelsen KK, Kankam M, Lund MT, Rubino DM, Satylganova A, et al. 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) 397:1736–48. doi: 10.1016/S0140-6736(21)00845-X

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Jastreboff AM, Kaplan LM, Frias JP, Wu Q, Du Y, Gurbuz S, et al. Triple-hormone-receptor agonist retatrutide for obesity - A phase 2 trial. N Engl J Med. (2023) 389:514–26. doi: 10.1056/NEJMoa2301972

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Blundell JE, Dulloo AG, Salvador J, Fruhbeck G. BMI ESWGo. Beyond BMI–phenotyping the obesities. Obes Facts. (2014) 7:322–8. doi: 10.1159/000368783

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Amaro A, Sugimoto D, Wharton S. Efficacy and safety of semaglutide for weight management: evidence from the STEP program. Postgrad Med. (2022) 134:5–17. doi: 10.1080/00325481.2022.2147326

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Wharton S, Lau DCW, Vallis M, Sharma AM, Biertho L, Campbell-Scherer 0D, et al. Obesity in adults: a clinical practice guideline. CMAJ. (2020) 192:E875–E91. doi: 10.1503/cmaj.191707

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Rubino DM, Greenway FL, Khalid U, O' Neil PM, Rosenstock J, Sorrig R, 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) 327:138–50. doi: 10.1001/jama.2021.23619

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Wadden TA, Hollander P, Klein S, Niswender K, Woo V, Hale PM, et al. Weight maintenance and additional weight loss with liraglutide after low-calorie-diet-induced weight loss: the SCALE Maintenance randomized study. Int J Obes (Lond). (2013) 37:1443–51. doi: 10.1038/ijo.2013.120

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Wadden TA, Chao AM, Machineni S, Kushner R, Ard J, Srivastava G, et al. Tirzepatide after intensive lifestyle intervention in adults with overweight or obesity: the SURMOUNT-3 phase 3 trial. Nat Med. (2023) 29:2909–18. doi: 10.1038/s41591-023-02597-w

PubMed Abstract | CrossRef Full Text | Google Scholar

39. King WC, Hinerman AS, Belle SH, Wahed AS, Courcoulas AP. Comparison of the performance of common measures of weight regain after bariatric surgery for association with clinical outcomes. JAMA. (2018) 320:1560–9. doi: 10.1001/jama.2018.14433

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Miras AD, Perez-Pevida B, Aldhwayan M, Kamocka A, McGlone ER, Al-Najim W, et al. Adjunctive liraglutide treatment in patients with persistent or recurrent type 2 diabetes after metabolic surgery (GRAVITAS): a randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol. (2019) 7:549–59. doi: 10.1016/S2213-8587(19)30157-3

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Mok J, Adeleke MO, Brown A, Kamocka A, McGlone ER, Al-Najim W, et al. Safety and Efficacy of Liraglutide, 3.0 mg, Once Daily vs Placebo in Patients With Poor Weight Loss Following Metabolic Surgery: The BARI-OPTIMISE Randomized Clinical Trial. JAMA Surg. (2023). doi: 10.1001/jamasurg.2023.2930

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Wharton S, Kuk JL, Luszczynski M, Kamran E, Christensen RAG. Liraglutide 3.0 mg for the management of insufficient weight loss or excessive weight regain post-bariatric surgery. Clin Obes. (2019) 9:e12323. doi: 10.1111/cob.12323

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Suliman M, Buckley A, Al Tikriti A, Tan T, Le Roux CW, Lessan N, et al. Routine clinical use of liraglutide 3 mg for the treatment of obesity: Outcomes in non-surgical and bariatric surgery patients. Diabetes Obes Metab. (2019) 21:1498–501. doi: 10.1111/dom.13672

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Jensen AB, Renstrom F, Aczel S, Folie P, Biraima-Steinemann M, Beuschlein F, et al. Efficacy of the glucagon-like peptide-1 receptor agonists liraglutide and semaglutide for the treatment of weight regain after bariatric surgery: a retrospective observational study. Obes Surg. (2023) 33:1017–25. doi: 10.1007/s11695-023-06484-8

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Lautenbach A, Wernecke M, Huber TB, Stoll F, Wagner J, Meyhofer SM, et al. The potential of semaglutide once-weekly in patients without type 2 diabetes with weight regain or insufficient weight loss after bariatric surgery-a retrospective analysis. Obes Surg. (2022) 32:3280–8. doi: 10.1007/s11695-022-06211-9

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Murvelashvili N, Xie L, Schellinger JN, Mathew MS, Marroquin EM, Lingvay I, et al. Effectiveness of semaglutide versus liraglutide for treating post-metabolic and bariatric surgery weight recurrence. Obes (Silver Spring). (2023) 31:1280–9. doi: 10.1002/oby.23736

CrossRef Full Text | Google Scholar

47. Lucas E, Simmons O, Tchang B, Aronne L. Pharmacologic management of weight regain following bariatric surgery. Front Endocrinol (Lausanne). (2022) 13:1043595. doi: 10.3389/fendo.2022.1043595

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Imam A, Alim H, Binhussein M, Kabli A, Alhasnani H, Allehyani A, et al. Weight loss effect of GLP-1 RAs with endoscopic bariatric therapy and bariatric surgeries. J Endocr Soc. (2023) 7:bvad129. doi: 10.1210/jendso/bvad129

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Weiss F, Barbuti M, Carignani G, Calderone A, Santini F, Maremmani I, et al. Psychiatric aspects of obesity: A narrative review of pathophysiology and psychopathology. J Clin Med. (2020) 9(8):2344. doi: 10.3390/jcm9082344

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Christensen SM, Varney C, Gupta V, Wenz L, Bays HE. Stress, psychiatric disease, and obesity: An Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) 2022. Obes Pillars. (2022) 4:100041. doi: 10.1016/j.obpill.2022.100041

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Boughton CK, Murphy KG. Can neuropeptides treat obesity? A review of neuropeptides and their potential role in the treatment of obesity. Br J Pharmacol. (2013) 170:1333–48. doi: 10.1111/bph.12037

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Kushner RF, Fink-Jensen A, Frenkel O, McGowan B, Goldman B, Overvad M, et al. Efficacy and safety of semaglutide 2.4 mg according to antidepressant use at baseline: A post hoc subgroup analysis. Obes (Silver Spring). (2023)

Google Scholar

53. Prasad F, De R, Korann V, Chintoh AF, Remington G, Ebdrup BH, et al. Semaglutide for the treatment of antipsychotic-associated weight gain in patients not responding to metformin - a case series. Ther Adv Psychopharmacol. (2023) 13:20451253231165169. doi: 10.1177/20451253231165169

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Sass MR, Danielsen AA, Kohler-Forsberg O, Storgaard H, Knop FK, Nielsen MO, et al. Effect of the GLP-1 receptor agonist semaglutide on metabolic disturbances in clozapine-treated or olanzapine-treated patients with a schizophrenia spectrum disorder: study protocol of a placebo-controlled, randomised clinical trial (SemaPsychiatry). BMJ Open. (2023) 13:e068652. doi: 10.1136/bmjopen-2022-068652

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Wang W, Volkow ND, Berger NA, Davis PB, Kaelber DC, Xu R. Association of semaglutide with risk of suicidal ideation in a real-world cohort. Nat Med. (2024). doi: 10.1038/s41591-023-02672-2

CrossRef Full Text | Google Scholar

56. Bergmann NC, Davies MJ, Lingvay I, Knop FK. Semaglutide for the treatment of overweight and obesity: A review. Diabetes Obes Metab. (2023) 25:18–35. doi: 10.1111/dom.14863

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Cao C, Yang S, Zhou Z. GLP-1 receptor agonists and pancreatic safety concerns in type 2 diabetic patients: data from cardiovascular outcome trials. Endocrine. (2020) 68:518–25. doi: 10.1007/s12020-020-02223-6

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Abd El Aziz M, Cahyadi O, Meier JJ, Schmidt WE, Nauck MA. Incretin-based glucose-lowering medications and the risk of acute pancreatitis and Malignancies: a meta-analysis based on cardiovascular outcomes trials. Diabetes Obes Metab. (2020) 22:699–704. doi: 10.1111/dom.13924

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Smits MM, Van Raalte DH. Safety of semaglutide. Front Endocrinol (Lausanne). (2021) 12:645563. doi: 10.3389/fendo.2021.645563

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Nagendra L, Bg H, Sharma M, Dutta D. Semaglutide and cancer: A systematic review and meta-analysis. Diabetes Metab Syndr. (2023) 17:102834. doi: 10.1016/j.dsx.2023.102834

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Sodhi M, Rezaeianzadeh R, Kezouh A, Etminan M. Risk of gastrointestinal adverse events associated with glucagon-like peptide-1 receptor agonists for weight loss. JAMA. (2023). doi: 10.1001/jama.2023.19574

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Wing RR, Look ARG. Does lifestyle intervention improve health of adults with overweight/obesity and type 2 diabetes? Findings from the look AHEAD randomized trial. Obes (Silver Spring). (2021) 29:1246–58. doi: 10.1002/oby.23158

CrossRef Full Text | Google Scholar

63. Ryan DH. Semaglutide for obesity: four STEPs forward, but more to come. Lancet Diabetes Endocrinol. (2021) 9:252–4. doi: 10.1016/S2213-8587(21)00081-4

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Heymsfield SB, Coleman LA, Miller R, Rooks DS, Laurent D, Petricoul O, 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. doi: 10.1001/jamanetworkopen.2020.33457

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: obesity, semaglutide, GLP 1 analog, overweight, anti-obesity medications

Citation: Tzoulis P and Baldeweg SE (2024) Semaglutide for weight loss: unanswered questions. Front. Endocrinol. 15:1382814. doi: 10.3389/fendo.2024.1382814

Received: 06 February 2024; Accepted: 22 May 2024;
Published: 05 June 2024.

Edited by:

Xiaodong Sun, Affiliated Hospital Of Shandong Second Medical University, China

Reviewed by:

Jean Albert Boutin, Institut de Recherche International Servier, France

Copyright © 2024 Tzoulis and Baldeweg. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ploutarchos Tzoulis, ptzoulis@yahoo.co.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.