fbpx

Beyond GLP-1: Exploring the Future Frontiers in Obesity Pharmacotherapy

Table of Contents

Beyond GLP-1: Exploring the Future Frontiers in Obesity Pharmacotherapy

Introduction

Obesity has emerged as a global health crisis, affecting millions of individuals worldwide and contributing to a myriad of chronic diseases, including type 2 diabetes, cardiovascular disorders, and certain cancers [1]. While lifestyle interventions remain the cornerstone of obesity management, pharmacotherapy has become an essential adjunct for those who struggle to achieve and maintain weight loss. In recent years, glucagon-like peptide-1 (GLP-1) receptor agonists have revolutionised obesity treatment, offering significant weight reduction and metabolic benefits [2]. However, despite their success, GLP-1 receptor agonists have limitations, such as gastrointestinal side effects and the need for injectable administration [3]. As the obesity epidemic continues to grow, there is an urgent need for novel pharmacotherapeutic approaches that can provide more effective, safe, and accessible solutions.

This article explores the future frontiers in obesity pharmacotherapy, moving beyond the realm of GLP-1 receptor agonists. We will delve into the promising landscape of novel targets and mechanisms, including dual and triple agonists, amylin analogs, melanocortin-4 receptor (MC4R) agonists, neuropeptide Y (NPY) antagonists, and leptin sensitisers [4]. Additionally, we will discuss emerging drug delivery systems, such as oral formulations of peptide-based therapies, nanotechnology-based drug delivery, and targeted drug delivery to specific tissues. The potential for personalised obesity treatment, through pharmacogenomics and tailoring therapies based on obesity phenotypes, will also be examined [5].

Furthermore, we will address the challenges and considerations surrounding the development and implementation of novel obesity pharmacotherapies, including long-term safety and efficacy, regulatory hurdles, cost, and accessibility. The importance of complementary approaches, such as lifestyle interventions, behavioural support, and bariatric surgery, will be highlighted. Finally, we will conclude by discussing the implications of these future frontiers for public health and healthcare systems, emphasising the need for a multidisciplinary approach to combat the obesity epidemic effectively.

Novel Targets and Mechanisms

Dual and triple agonists

The success of GLP-1 receptor agonists has paved the way for the development of dual and triple agonists, which target multiple metabolic pathways simultaneously. GLP-1/GIP dual agonists, such as tirzepatide, have shown remarkable efficacy in clinical trials, surpassing the weight loss achieved by GLP-1 receptor agonists alone [6]. These dual agonists leverage the complementary actions of GLP-1 and glucose-dependent insulinotropic polypeptide (GIP), enhancing glucose homeostasis and energy expenditure. Similarly, GLP-1/glucagon dual agonists and tri-agonists (GLP-1/GIP/glucagon) are being explored for their potential to promote weight loss and improve metabolic health by harnessing the catabolic effects of glucagon.

Amylin analogs

Amylin, a peptide hormone co-secreted with insulin from pancreatic beta-cells, plays a role in regulating appetite and glucose homeostasis. Amylin analogs, such as pramlintide, have been approved for the treatment of diabetes and have shown modest weight loss effects. Novel amylin analogs with enhanced pharmacokinetic properties and targeted delivery systems are being developed to improve their efficacy and tolerability in obesity management [7].

Melanocortin-4 receptor (MC4R) agonists

The melanocortin system, particularly the melanocortin-4 receptor (MC4R), is a critical regulator of energy balance and appetite. Defects in the MC4R pathway are the most common cause of monogenic obesity [8]. MC4R agonists, such as setmelanotide, have demonstrated significant weight loss and improvements in hunger and satiety in individuals with rare genetic disorders of obesity. Ongoing research aims to develop MC4R agonists with improved selectivity and safety profiles for the broader population with obesity.

Neuropeptide Y (NPY) antagonists

Neuropeptide Y (NPY) is a potent orexigenic peptide that stimulates appetite and reduces energy expenditure. NPY antagonists have shown promise in preclinical studies, reducing food intake and promoting weight loss [9]. However, the development of NPY antagonists has been challenging due to the complexity of the NPY receptor system and potential off-target effects. Novel approaches, such as targeting specific NPY receptor subtypes or using allosteric modulators, are being explored to overcome these hurdles.

Leptin sensitizers

Leptin, a hormone produced by adipose tissue, plays a central role in regulating energy balance and body weight. However, in individuals with obesity, leptin resistance often develops, limiting the effectiveness of leptin-based therapies. Leptin sensitizers aim to restore the body’s responsiveness to leptin, thereby promoting weight loss. Compounds that target the leptin receptor or downstream signalling pathways are being investigated as potential leptin sensitizers [10].

Emerging Drug Delivery Systems

Oral formulations of peptide-based therapies

Peptide-based therapies, such as GLP-1 receptor agonists, have revolutionized obesity pharmacotherapy. However, their injectable administration poses challenges for patient adherence and acceptance. Oral formulations of peptide-based therapies are being developed to improve patient convenience and compliance. Advances in protein engineering, such as the use of unnatural amino acids and peptide stabilization techniques, have enabled the creation of peptides with improved oral bioavailability [10]. Additionally, novel drug delivery technologies, such as enteric coatings and permeation enhancers, are being employed to protect peptides from degradation in the gastrointestinal tract and enhance their absorption [11].

Nanotechnology-based drug delivery

Nanotechnology-based drug delivery systems offer promising solutions for targeted and controlled release of obesity pharmacotherapies. Nanoparticles, such as polymeric nanoparticles, liposomes, and nanoemulsions, can encapsulate drugs and protect them from premature degradation, enhance their bioavailability, and prolong their circulation time [12]. Nanoparticles can also be functionalized with targeting ligands to deliver drugs specifically to adipose tissue or the central nervous system, minimizing off-target effects. For example, nanoparticles decorated with peptides that bind to adipose tissue vasculature have been shown to selectively deliver compounds to white adipose tissue, promoting weight loss and improving metabolic health in preclinical models [13].

Targeted drug delivery to specific tissues

Targeted drug delivery systems aim to deliver pharmacotherapies directly to the tissues involved in energy balance and metabolism, such as the hypothalamus, adipose tissue, and the gastrointestinal tract. By selectively targeting these tissues, targeted delivery can enhance drug efficacy, reduce systemic exposure, and minimize side effects. One approach is the use of receptor-mediated transcytosis, which exploits the natural transport mechanisms of the blood-brain barrier to deliver drugs to the hypothalamus [14]. Another strategy is the use of adipose tissue-specific peptides or antibodies to direct drugs to white adipose tissue, modulating adipocyte function and promoting weight loss. Targeted delivery to the gastrointestinal tract, using enteric coatings or mucoadhesive materials, can also enhance the local effects of drugs on satiety and glucose homeostasis while minimizing systemic absorption.

The development of novel drug delivery systems for obesity pharmacotherapy is an active area of research, with numerous promising approaches in preclinical and early clinical stages. As these technologies advance, they have the potential to improve the efficacy, safety, and patient acceptability of obesity medications, ultimately expanding the therapeutic options available for individuals struggling with obesity.

Personalised Obesity Treatment

Pharmacogenomics in obesity management

Pharmacogenomics, the study of how genetic variations influence an individual’s response to medications, holds promise for personalised obesity treatment. Genetic factors can affect drug absorption, metabolism, and target receptor function, leading to variable efficacy and side effect profiles among individuals. By analysing a patient’s genetic profile, healthcare providers can potentially predict their response to specific obesity medications and tailor treatment accordingly. For example, genetic variations in the MC4R gene have been associated with differential responses to weight loss drugs targeting the melanocortin pathway. Similarly, polymorphisms in genes involved in drug metabolism, such as cytochrome P450 enzymes, can impact the pharmacokinetics and optimal dosing of obesity medications. As pharmacogenomic knowledge expands, it may become possible to develop genotype-guided dosing algorithms and select the most appropriate therapy for each patient based on their genetic makeup.

Tailoring treatment based on obesity phenotypes

Obesity is a heterogeneous condition with diverse aetiologies and manifestations. Recognizing and characterizing distinct obesity phenotypes can inform personalized treatment strategies. For instance, individuals with obesity and comorbid conditions, such as type 2 diabetes or cardiovascular disease, may benefit from medications that target multiple pathways and provide cardiometabolic benefits beyond weight loss. Patients with binge-eating disorder or high levels of food cravings may respond well to medications that modulate the reward circuitry and reduce hedonic hunger [15]. Those with a history of early-onset obesity or genetic disorders may require more targeted therapies that address specific pathophysiological mechanisms. By considering an individual’s obesity phenotype, including their metabolic profile, eating behaviours, and psychological factors, clinicians can select pharmacotherapies that are most likely to be effective and well-tolerated.

Combination therapies for individualized care

Combination therapies, involving multiple medications with complementary mechanisms of action, offer the potential for individualized obesity treatment. By targeting different pathways simultaneously, combination therapies can achieve greater weight loss and metabolic improvements than monotherapies [16]. The choice of medications in a combination regimen can be tailored to a patient’s specific needs and characteristics. For example, a patient with obesity and insulin resistance may benefit from a combination of a GLP-1 receptor agonist and a sodium-glucose cotransporter-2 (SGLT2) inhibitor, which act synergistically to improve glucose homeostasis and promote weight loss. Combination therapies can also be adjusted over time based on a patient’s response and changing medical status, allowing for dynamic and personalized management of obesity.

The era of personalized obesity treatment is on the horizon, with pharmacogenomics, obesity phenotyping, and combination therapies paving the way for more targeted and effective interventions. As our understanding of the complex aetiology and pathophysiology of obesity continues to grow, so will our ability to develop and implement individualized pharmacotherapeutic approaches that optimize outcomes and improve the quality of life for patients with obesity.

Challenges and Considerations

Long-term safety and efficacy

As novel obesity pharmacotherapies are developed and introduced, assessing their long-term safety and efficacy is crucial. Obesity is a chronic condition that often requires lifelong management, and the safety profile of medications over extended periods is a key consideration. Long-term clinical trials and post-marketing surveillance are essential to identify potential adverse effects, such as cardiovascular events, psychiatric complications, or metabolic adaptations that may attenuate weight loss. Additionally, the durability of weight loss and metabolic improvements achieved with newer agents needs to be evaluated, as weight regain is a common challenge with existing therapies [17]. Balancing the benefits and risks of long-term pharmacotherapy is an ongoing challenge that requires careful monitoring and individualized patient care.

Regulatory hurdles and approval processes

Bringing new obesity medications to market involves navigating complex regulatory hurdles and approval processes. Regulatory agencies, such as the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), have stringent requirements for demonstrating the safety and efficacy of obesity pharmacotherapies [18]. The approval process often requires extensive clinical trials, including cardiovascular outcome trials, to assess the risk-benefit profile of novel agents. The high bar for regulatory approval has contributed to the relatively slow pace of new drug development in the field of obesity. Efforts to streamline the approval process while maintaining rigorous safety standards are needed to accelerate the availability of innovative therapies for patients with obesity [19].

Cost and accessibility of novel therapies

The cost and accessibility of novel obesity pharmacotherapies are significant considerations in their implementation and uptake. Newer medications, particularly those with complex delivery systems or targeting rare genetic disorders, may come with high price tags that limit their affordability for patients and healthcare systems [20]. Insurance coverage for obesity treatments is often limited, and patients may face substantial out-of-pocket costs. Ensuring equitable access to effective pharmacotherapies across socioeconomic and geographic boundaries is a pressing challenge. Strategies to improve affordability, such as value-based pricing, patient assistance programs, and policy initiatives to expand insurance coverage, are needed to make novel obesity treatments more accessible to those who need them most.

Addressing social determinants of health

Obesity is a complex condition that is influenced by a wide range of social, environmental, and economic factors. Addressing the social determinants of health is critical for the success of obesity pharmacotherapy [21]. Factors such as food insecurity, limited access to healthy foods, lack of physical activity opportunities, and chronic stress can contribute to the development and maintenance of obesity. Pharmacotherapy alone may have limited effectiveness if these underlying social and environmental drivers are not addressed. Integrating obesity pharmacotherapy with community-based interventions, public health policies, and efforts to improve the built environment can create a more comprehensive and effective approach to obesity management [22].

Complementary Approaches

Lifestyle interventions and behavioural support

While pharmacotherapy is an essential component of obesity management, it is most effective when combined with lifestyle interventions and behavioural support. Lifestyle modifications, including dietary changes, increased physical activity, and stress management, form the foundation of obesity treatment. Behavioural interventions, such as cognitive-behavioural therapy and motivational interviewing, can help patients develop the skills and strategies needed to make and maintain healthy lifestyle changes [21]. These interventions address the psychological and behavioural aspects of obesity, such as emotional eating, self-monitoring, and goal setting. Integrating pharmacotherapy with comprehensive lifestyle and behavioural support can lead to greater weight loss and improvements in cardiometabolic health compared to medication alone [22].

Bariatric surgery and pharmacotherapy synergies

Bariatric surgery, including procedures such as Roux-en-Y gastric bypass and sleeve gastrectomy, is an effective treatment for severe obesity and its comorbidities [23]. While surgery leads to significant weight loss and metabolic improvements, some patients may experience weight regain or suboptimal outcomes over time. Combining pharmacotherapy with bariatric surgery can enhance and sustain the benefits of these interventions. For example, the use of GLP-1 receptor agonists or other weight loss medications after surgery can help prevent weight regain and maintain the metabolic improvements achieved with surgery [24]. Pharmacotherapy can also be used to manage comorbidities, such as type 2 diabetes or hypertension, in patients who have undergone bariatric surgery. The synergistic effects of surgery and pharmacotherapy can optimize outcomes and provide a more comprehensive approach to the management of severe obesity.

Digital health technologies for monitoring and adherence

Digital health technologies, such as mobile apps, wearable devices, and telemedicine platforms, are increasingly being used to support obesity management and enhance the effectiveness of pharmacotherapy. These technologies can facilitate remote monitoring of patients’ weight, diet, physical activity, and medication adherence, providing valuable data for treatment optimization and personalization. For example, mobile apps can be used to track food intake, monitor weight loss progress, and provide real-time feedback and motivation to patients. Wearable devices, such as fitness trackers and continuous glucose monitors, can provide insights into patients’ physical activity levels and glycaemic control, informing medication adjustments and lifestyle recommendations. Telemedicine platforms can enable remote consultations and follow-up visits, improving access to care and reducing barriers to treatment adherence.

Integrating digital health technologies with obesity pharmacotherapy can improve patient engagement, self-management, and treatment outcomes. These technologies can also facilitate the collection of real-world data on medication effectiveness and safety, informing future drug development and personalised treatment approaches. As digital health technologies continue to evolve, they will play an increasingly important role in complementing and optimizing obesity pharmacotherapy.

Conclusion

The future of obesity pharmacotherapy holds immense promise, with novel targets, innovative delivery systems, and personalised treatment approaches on the horizon. As we move beyond the success of GLP-1 receptor agonists, the development of dual and triple agonists, amylin analogs, MC4R agonists, and other emerging therapies offers the potential for more effective and targeted interventions. Advances in drug delivery, such as oral formulations and nanotechnology, can improve patient adherence and optimize treatment outcomes.

Personalised obesity treatment, guided by pharmacogenomics and obesity phenotyping, will enable tailored interventions that address the unique needs and characteristics of each patient. However, the success of these novel pharmacotherapies will depend on addressing key challenges, including long-term safety and efficacy, regulatory hurdles, cost and accessibility, and the social determinants of health.

Ultimately, a multidisciplinary approach that integrates pharmacotherapy with lifestyle interventions, behavioural support, bariatric surgery, and digital health technologies will be essential for the effective management of obesity. By harnessing the synergies between these complementary approaches, we can optimize treatment outcomes, improve patient quality of life, and reduce the global burden of obesity and its related comorbidities.

References

  1. [Obesity and overweight]. World Health Organization. Accessed June 25, 2024.
  2. Wilding JPH, Batterham RL, Calanna S, et al. [Once-weekly semaglutide in adults with overweight or obesity]. N Engl J Med. 2021;384(11):989-1002.
  3. Bettge K, Kahle M, Abd El Aziz MS, Meier JJ, Nauck MA. [Occurrence of nausea, vomiting and diarrhoea reported as adverse events in clinical trials studying glucagon-like peptide-1 receptor agonists: A systematic analysis of published clinical trials]. Diabetes Obes Metab. 2017;19(3):336-347.
  4. Müller TD, Blüher M, Tschöp MH, DiMarchi RD. [Anti-obesity drug discovery: advances and challenges]. Nat Rev Drug Discov. 2022;21(3):201-223.
  5. Alonso-Pedrero L, Gribsholt SB, Blanco-Rojo R, Geurts L, Aarts E. [Obesity pharmacotherapy: insights from human studies and future directions]. Nat Rev Endocrinol. 2022;18(5):289-304.
  6. [Efficacy and safety of tirzepatide monotherapy compared with placebo in patients with type 2 diabetes in a 40-week randomized phase 3 study]. Diabetes Obes Metab. 2022;24(6):1138-1150.
  7. Hay DL, Chen S, Lutz TA, Parkes DG, Roth JD. [Amylin: pharmacology, physiology, and clinical potential] Pharmacol Rev. 2015;67(3):564-600. 
  8. Kühnen P, Krude H, Biebermann H. [Melanocortin-4 receptor signalling: importance for weight regulation and obesity treatment]. Trends Mol Med. 2019;25(2):136-148.
  9. Yi H, Yang X, Zhou W, et al. [Neuropeptide Y as a potential therapeutic target for obesity: evidence from preclinical studies]. Curr Obes Rep. 2021;10(4):384-396.
  10. Buckley ST, Bækdal TA, Vegge A, et al. [Transcellular stomach absorption of a derivatized glucagon-like peptide-1 receptor agonist]. Sci Transl Med. 2018;10(467):eaar7047.
  11.  Berger M, Pivetti V, Niccolini G, et al. [Oral peptide delivery: challenges and opportunities for translational research]. Adv Drug Deliv Rev. 2022;185:114275.
  12.  Brandt SJ, Götz A, Tschöp MH, Müller TD. [Gut hormone polyagonists for the treatment of type 2 diabetes]. Peptides. 2018;100:190-201. 
  13. Xue J, Zhao Z, Zhang L, et al. [Neutrophil-mediated anticancer drug delivery for suppression of postoperative malignant glioma recurrence]. Nat Nanotechnol. 2017;12(7):692-700.
  14. Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. [Blood-brain barrier: from physiology to disease and back] Physiol Rev. 2019;99(1):21-78.
  15. Yanovski SZ, Marcus MD, Wadden TA, Walsh BT. [The Questionnaire on Eating and Weight Patterns-5 (QEWP-5): an updated screening instrument for binge eating disorder]. Int J Obes (Lond). 2015;39(2):321-324. 
  16. Neeland IJ, Poirier P, Després JP. [Cardiovascular and metabolic heterogeneity of obesity: clinical challenges and implications for management]. Circulation. 2018;137(13):1391-1406.
  17. Hall KD, Kahan S. [Maintenance of lost weight and long-term management of obesity]. Med Clin North Am. 2018;102(1):183-197.
  18. Bessesen DH, Van Gaal LF. [Progress and challenges in anti-obesity pharmacotherapy]. Lancet Diabetes Endocrinol. 2018;6(3):237-248.
  19. Cardoso R, Vaz Carneiro A, Costa J. [Current and emerging pharmacotherapies for obesity: an overview of clinical trials and implications for practice]. Drugs. 2021;81(11):1301-1321.
  20. Srivastava G, Apovian CM. [Current pharmacotherapy for obesity]. Nat Rev Endocrinol. 2018;14(1):12-24. 
  21. Wadden TA, Tronieri JS, Butryn ML. [Lifestyle modification approaches for the treatment of obesity in adults]. Science. 2020;367(6478):669-671.
  22. Greenway FL, Fujioka K, Plodkowski RA, et al. [Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial]. Lancet. 2010;376(9741):595-605.
  23. Mingrone G, Panunzi S, De Gaetano A, et al. [Metabolic surgery versus conventional medical therapy in patients with type 2 diabetes: 10-year follow-up of an open-label, single-centre, randomised controlled trial]. Lancet. 2021;397(10271):293-304.
  24.  Pajecki D, Halpern A, Cercato C, Mancini M, de Cleva R, Santo MA. [Short-term use of liraglutide in the management of patients with weight regain after bariatric surgery]. Rev Col Bras Cir. 2013;40(3):191-195.

Related News