1
|
Wean J, Kowalsky AH, Laker R, Will S, Drucker DJ, Rhodes CJ, Seeley RJ. Specific loss of GIPR signaling in GABAergic neurons enhances GLP-1R agonist-induced body weight loss. Mol Metab 2025; 95:102074. [PMID: 39612941 PMCID: PMC11946504 DOI: 10.1016/j.molmet.2024.102074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
OBJECTIVES Dual incretin agonists are among the most effective pharmaceutical treatments for obesity and type 2 diabetes to date. Such therapeutics can target two receptors, such as the glucagon-like peptide-1 (GLP-1) receptor and the glucose-dependent insulinotropic polypeptide (GIP) receptor in the case of tirzepatide, to improve glycemia and reduce body weight. Regarding body weight effects, GIPR signaling is thought to involve at least two relevant mechanisms: the enhancement of food intake reduction and the attenuation of aversive effects caused by GLP-1R agonists. Although it is known that dual GLP-1R-GIPR agonism produces greater weight loss than GLP-1R agonism alone, the precise mechanism is unknown. METHODS To address this question, we used mice lacking GIPR in the whole body, GABAergic neurons, or glutamatergic neurons. These mice were given various combinations of GLP-1R and GIPR agonist drugs with subsequent food intake and conditioned taste aversion measurements. RESULTS A GIPR knockout in either the whole body or selectively in inhibitory GABAergic neurons protects against diet-induced obesity, whereas a knockout in excitatory glutamatergic neurons had a negligible effect. Furthermore, we found that GIPR in GABAergic neurons is essential for the enhanced weight loss efficacy of dual incretin agonism, yet, surprisingly, its removal enhances the effect of GLP-1R agonism alone. Finally, GIPR knockout in GABAergic neurons prevents the anti-aversive effects of GIPR agonism. CONCLUSIONS Our findings are consistent with GIPR research at large in that both enhancement and removal of GIPR signaling are metabolically beneficial. Notably, however, our findings suggest that future obesity therapies designed to modulate GIPR signaling, whether by agonism or antagonism, would be best targeted towards GABAergic neurons.
Collapse
Affiliation(s)
- Jordan Wean
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Rhianna Laker
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Sarah Will
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Daniel J Drucker
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Department of Medicine, University of Toronto, Toronto, Canada
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Bailey CJ, Flatt PR, Conlon JM. Multifunctional incretin peptides in therapies for type 2 diabetes, obesity and associated co-morbidities. Peptides 2025; 187:171380. [PMID: 40081498 DOI: 10.1016/j.peptides.2025.171380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Recent studies with peptide-based incretin therapies have focussed mainly on the glucagon-like peptide-1 (GLP-1) receptor agonist semaglutide and the dual agonist tirzepatide that engages receptors for GLP-1 and glucose-dependent insulinotropic polypeptide (GIP). Randomised clinical trials and 'real-world' studies have confirmed the marked glucose-lowering and weight-lowering efficacy of these agents across diverse populations. These include different ethnic groups, young and elderly individuals with and without diabetes and/or overweight or obesity. Recent studies have also confirmed protections against the development and progression of cardiovascular and renal diseases that are additive to the benefits conferred by improved control of blood glucose and body weight. Emerging evidence suggests that incretin therapies could additionally ameliorate fatty liver disease, chronic inflammation, sleep apnea and possibly degenerative bone disorders and cognitive decline. New incretin-based peptide therapies in development include a long-acting glucagon receptor agonist (LY3324954), dual GLP-1/glucagon receptor agonists (survodutide, pemvidutide, mazdutide, G49), triple GLP-1/GIP/glucagon receptor agonists (retatrutide, efocipegtrutide), a combination of semaglutide with the amylin analogue cagrilintide (CagriSema), a unimolecular GLP-1/amylin receptor dual agonist (amycretin), and a GIP receptor antibody with GLP-1 receptor agonism (MariTide). The creation of multi-targeting incretin-based synthetic peptides provides opportunities for improved management of type 2 diabetes and obesity as well as new therapeutic approaches to an expanding list of associated co-morbidities. The aim of the review is to acquaint the reader with developments in the field from 2023 to the present (February 2025).
Collapse
Affiliation(s)
| | - Peter R Flatt
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - J Michael Conlon
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK.
| |
Collapse
|
3
|
Müller TD, Adriaenssens A, Ahrén B, Blüher M, Birkenfeld AL, Campbell JE, Coghlan MP, D'Alessio D, Deacon CF, DelPrato S, Douros JD, Drucker DJ, Figueredo Burgos NS, Flatt PR, Finan B, Gimeno RE, Gribble FM, Hayes MR, Hölscher C, Holst JJ, Knerr PJ, Knop FK, Kusminski CM, Liskiewicz A, Mabilleau G, Mowery SA, Nauck MA, Novikoff A, Reimann F, Roberts AG, Rosenkilde MM, Samms RJ, Scherer PE, Seeley RJ, Sloop KW, Wolfrum C, Wootten D, DiMarchi RD, Tschöp MH. Glucose-dependent insulinotropic polypeptide (GIP). Mol Metab 2025; 95:102118. [PMID: 40024571 PMCID: PMC11931254 DOI: 10.1016/j.molmet.2025.102118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glucose-dependent insulinotropic polypeptide (GIP) was the first incretin identified and plays an essential role in the maintenance of glucose tolerance in healthy humans. Until recently GIP had not been developed as a therapeutic and thus has been overshadowed by the other incretin, glucagon-like peptide 1 (GLP-1), which is the basis for several successful drugs to treat diabetes and obesity. However, there has been a rekindling of interest in GIP biology in recent years, in great part due to pharmacology demonstrating that both GIPR agonism and antagonism may be beneficial in treating obesity and diabetes. This apparent paradox has reinvigorated the field, led to new lines of investigation, and deeper understanding of GIP. SCOPE OF REVIEW In this review, we provide a detailed overview on the multifaceted nature of GIP biology and discuss the therapeutic implications of GIPR signal modification on various diseases. MAJOR CONCLUSIONS Following its classification as an incretin hormone, GIP has emerged as a pleiotropic hormone with a variety of metabolic effects outside the endocrine pancreas. The numerous beneficial effects of GIPR signal modification render the peptide an interesting candidate for the development of pharmacotherapies to treat obesity, diabetes, drug-induced nausea and both bone and neurodegenerative disorders.
Collapse
Affiliation(s)
- Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany; Walther-Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University Munich (LMU), Germany.
| | - Alice Adriaenssens
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Bo Ahrén
- Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Matthias Blüher
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen 72076, Germany; Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich, Tübingen, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Department of Medicine, Division of Endocrinology, Duke University, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Matthew P Coghlan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - David D'Alessio
- Department of Medicine, Division of Endocrinology, Duke University, Durham, NC, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Carolyn F Deacon
- School of Biomedical Sciences, Ulster University, Coleraine, UK; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefano DelPrato
- Interdisciplinary Research Center "Health Science", Sant'Anna School of Advanced Studies, Pisa, Italy
| | | | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Natalie S Figueredo Burgos
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Peter R Flatt
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Brian Finan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Ruth E Gimeno
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Fiona M Gribble
- Institute of Metabolic Science-Metabolic Research Laboratories & MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Matthew R Hayes
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christian Hölscher
- Neurodegeneration Research Group, Henan Academy of Innovations in Medical Science, Xinzheng, China
| | - Jens J Holst
- Department of Biomedical Sciences and the Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Patrick J Knerr
- Indianapolis Biosciences Research Institute, Indianapolis, IN, USA
| | - Filip K Knop
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christine M Kusminski
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany; Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Guillaume Mabilleau
- Univ Angers, Nantes Université, ONIRIS, Inserm, RMeS UMR 1229, Angers, France; CHU Angers, Departement de Pathologie Cellulaire et Tissulaire, Angers, France
| | | | - Michael A Nauck
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Aaron Novikoff
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany
| | - Frank Reimann
- Institute of Metabolic Science-Metabolic Research Laboratories & MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Anna G Roberts
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences University of Copenhagen, Copenhagen, Denmark
| | - Ricardo J Samms
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Philip E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kyle W Sloop
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Denise Wootten
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia; ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | | | - Matthias H Tschöp
- Helmholtz Munich, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
4
|
Roberts TD, Hutchinson DS, Wootten D, De Blasio MJ, Ritchie RH. Advances in incretin therapies for targeting cardiovascular disease in diabetes. J Mol Cell Cardiol 2025; 202:102-115. [PMID: 40086589 DOI: 10.1016/j.yjmcc.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
The global prevalence of obesity is skyrocketing at an alarming rate, with recent data estimating that one-in-eight people are now living with the disease. Obesity is a chronic metabolic disorder that shares underlying pathophysiology with other metabolically-linked diseases such as type 2 diabetes mellitus, cardiovascular disease and diabetic cardiomyopathy. There is a distinct correlation between type 2 diabetes status and the likelihood of heart failure. Of note, there is an apparent sexual dimorphism, with women disproportionately affected with respect to the degree of severity of the cardiac phenotype of diabetic cardiomyopathy that results from diabetes. The current pharmacotherapies available for the attenuation of hyperglycaemia in type 2 diabetes are not always effective, and have varying degrees of efficacy in the setting of heart failure. Insulin can worsen heart failure prognosis whereas metformin, sodium-glucose cotransporter 2 inhibitors (SGLT2i) and more recently, glucagon-like peptide-1 receptor agonists (GLP-1RAs), have demonstrated cardioprotection with their administration. This review will highlight the advancement of incretin therapies for individuals with diabetes and heart failure and explore newly-reported evidence of the clinical usefulness of GLP-1R agonists in this distinct phenotype of heart failure.
Collapse
Affiliation(s)
- Timothy D Roberts
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Dana S Hutchinson
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia
| | - Denise Wootten
- Metabolic G Protein-Coupled Receptor Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia; ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Miles J De Blasio
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| | - Rebecca H Ritchie
- Heart Failure Pharmacology Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, VIC, Australia.
| |
Collapse
|
5
|
Adriaenssens AE. Unravelling the GIPR agonist versus antagonist paradox. Nat Metab 2025:10.1038/s42255-025-01299-6. [PMID: 40301584 DOI: 10.1038/s42255-025-01299-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Affiliation(s)
- Alice E Adriaenssens
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| |
Collapse
|
6
|
Sedrak P, Verma R, Verma M, Connelly KA. Evolving Role of Double and Triple Therapy With GLP-1 Receptor Agonists in Obesity and Cardiovascular Disease. Can J Cardiol 2025:S0828-282X(25)00326-5. [PMID: 40311673 DOI: 10.1016/j.cjca.2025.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/18/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Glucagon-like peptide 1 receptor agonists (GLP-1RAs) have emerged as a transformative class of therapies, expanding their clinical utility far beyond glycemic control. Initially developed for the treatment of diabetes, these agents are now recognised as potent therapies for managing overweight and obesity, atherosclerosis, and heart failure. This review explores the evolution of GLP-1RA-based therapies, with a focus on novel advances such as dual GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) receptor agonists ("double G") and triple receptor agonists incorporating glucagon modulation ("triple G"). We also provide an overview of completed and ongoing clinical trials investigating the role of GLP-1RAs in atherosclerosis and heart failure. These developments underscore the expanding therapeutic landscape of GLP-1RAs and their growing significance in cardiometabolic medicine.
Collapse
Affiliation(s)
- Phelopater Sedrak
- Department of Internal Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Raj Verma
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Meena Verma
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kim A Connelly
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Division of Cardiology, Li Ka Shing Knowledge Institute of St Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Gutgesell RM, Khalil A, Liskiewicz A, Maity-Kumar G, Novikoff A, Grandl G, Liskiewicz D, Coupland C, Karaoglu E, Akindehin S, Castelino R, Curion F, Liu X, Garcia-Caceres C, Cebrian-Serrano A, Douros JD, Knerr PJ, Finan B, DiMarchi RD, Sloop KW, Samms RJ, Theis FJ, Tschöp MH, Müller TD. GIPR agonism and antagonism decrease body weight and food intake via different mechanisms in male mice. Nat Metab 2025:10.1038/s42255-025-01294-x. [PMID: 40301583 DOI: 10.1038/s42255-025-01294-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/28/2025] [Indexed: 05/01/2025]
Abstract
Agonists and antagonists of the glucose-dependent insulinotropic polypeptide receptor (GIPR) enhance body weight loss induced by glucagon-like peptide-1 receptor (GLP-1R) agonism. However, while GIPR agonism decreases body weight and food intake in a GLP-1R-independent manner via GABAergic GIPR+ neurons, it remains unclear whether GIPR antagonism affects energy metabolism via a similar mechanism. Here we show that the body weight and food intake effects of GIPR antagonism are eliminated in mice with global loss of either Gipr or Glp-1r but are preserved in mice with loss of Gipr in either GABAergic neurons of the central nervous system or peripherin-expressing neurons of the peripheral nervous system. Single-nucleus RNA-sequencing shows opposing effects of GIPR agonism and antagonism in the dorsal vagal complex, with antagonism, but not agonism, closely resembling GLP-1R signalling. Additionally, GIPR antagonism and GLP-1R agonism both regulate genes implicated in synaptic plasticity. Collectively, we show that GIPR agonism and antagonism decrease body weight via different mechanisms, with GIPR antagonism, unlike agonism, depending on functional GLP-1R signalling.
Collapse
Affiliation(s)
- Robert M Gutgesell
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Munich, Munich, Germany
| | - Ahmed Khalil
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
| | - Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Gandhari Maity-Kumar
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
| | - Aaron Novikoff
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
| | - Gerald Grandl
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
| | - Daniela Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Callum Coupland
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
| | - Ezgi Karaoglu
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, Eberhard Karls University, Tübingen, Germany
| | - Seun Akindehin
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
| | - Russell Castelino
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
| | - Fabiola Curion
- Department of Computational Health, Institute of Computational Biology, Helmholtz, Munich, Germany
- Department of Mathematics, School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Xue Liu
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
| | - Cristina Garcia-Caceres
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians Universität München, Munich, Germany
| | - Alberto Cebrian-Serrano
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany
- German Center for Diabetes Research, DZD, Neuherberg, Germany
| | | | - Patrick J Knerr
- Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Brian Finan
- Diabetes, Obesity and Complications Therapeutic Area, Eli Lilly and Company, Indianapolis, IN, USA
| | - Richard D DiMarchi
- Department of Chemistry, Indiana University Bloomington, Bloomington, IN, USA
| | - Kyle W Sloop
- Diabetes, Obesity and Complications Therapeutic Area, Eli Lilly and Company, Indianapolis, IN, USA
| | - Ricardo J Samms
- Diabetes, Obesity and Complications Therapeutic Area, Eli Lilly and Company, Indianapolis, IN, USA
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Munich, Munich, Germany
- Department of Mathematics, School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Matthias H Tschöp
- Helmholtz Munich, Munich, Germany.
- Division of Metabolic Diseases, Department of Medicine, Technische Universität, Munich, Germany.
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz, Munich, Germany.
- German Center for Diabetes Research, DZD, Neuherberg, Germany.
- Walther-Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians University Munich, Munich, Germany.
| |
Collapse
|
8
|
Liu CM, Killion EA, Hammoud R, Lu SC, Komorowski R, Liu T, Kanke M, Thomas VA, Cook K, Sivits GN, Ben AB, Atangan LI, Hussien R, Tang A, Shkumatov A, Li CM, Drucker DJ, Véniant MM. GIPR-Ab/GLP-1 peptide-antibody conjugate requires brain GIPR and GLP-1R for additive weight loss in obese mice. Nat Metab 2025:10.1038/s42255-025-01295-w. [PMID: 40301582 DOI: 10.1038/s42255-025-01295-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/28/2025] [Indexed: 05/01/2025]
Abstract
Glucose-dependent insulinotropic polypeptide receptor (GIPR) and glucagon-like peptide 1 receptor (GLP-1R) are expressed in the central nervous system (CNS) and regulate food intake. Here, we demonstrate that a peptide-antibody conjugate that blocks GIPR while simultaneously activating GLP-1R (GIPR-Ab/GLP-1) requires both CNS GIPR and CNS GLP-1R for maximal weight loss in obese, primarily male, mice. Moreover, dulaglutide produces greater weight loss in CNS GIPR knockout (KO) mice, and the weight loss achieved with dulaglutide + GIPR-Ab is attenuated in CNS GIPR KO mice. Wild-type mice treated with GIPR-Ab/GLP-1 and CNS GIPR KO mice exhibit similar changes in gene expression related to tissue remodelling, lipid metabolism and inflammation in white adipose tissue and liver. Moreover, GIPR-Ab/GLP-1 is detected in circumventricular organs in the brain and activates c-FOS in downstream neural substrates involved in appetite regulation. Hence, both CNS GIPR and GLP-1R signalling are required for the full weight loss effect of a GIPR-Ab/GLP-1 peptide-antibody conjugate.
Collapse
Affiliation(s)
- Clarissa M Liu
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
- Amgen R&D Postdoctoral Fellows Program, Amgen Inc., Thousand Oaks, CA, USA
| | - Elizabeth A Killion
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Rola Hammoud
- The Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Shu-Chen Lu
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Renee Komorowski
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Tongyu Liu
- Center for Research Acceleration by Digital Innovation, Amgen Research, Thousand Oaks, CA, USA
| | - Matt Kanke
- Department of Research Technologies, Amgen Research, South San Francisco, CA, USA
| | - Veena A Thomas
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Kevin Cook
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Glenn N Sivits
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Aerielle B Ben
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Larissa I Atangan
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Rajaa Hussien
- Department of Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Amy Tang
- Department of Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Artem Shkumatov
- Department of Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Chi-Ming Li
- Department of Research Technologies, Amgen Research, South San Francisco, CA, USA
| | - Daniel J Drucker
- The Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Murielle M Véniant
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA.
| |
Collapse
|
9
|
Zhou C, Gong B, Liu X, Hu G, Sun L. Glucose-dependent insulinotropic peptide and beyond: co-agonist innovations in the treatment of metabolic diseases. Eur J Pharmacol 2025; 999:177681. [PMID: 40306536 DOI: 10.1016/j.ejphar.2025.177681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 04/12/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025]
Abstract
Glucose-dependent insulinotropic peptide (GIP), a key incretin hormone, has emerged as a pivotal therapeutic target in metabolic disorders. Historically, its therapeutic potential in type 2 diabetes mellitus (T2DM) has been underestimated owing to GIP resistance and its limited acute effects on glycemic control and body weight regulation. However, emerging evidence has demonstrated that GIP resistance is reversible through sustained glycemic improvement, thereby restoring its physiological effectiveness. With the development of gut hormone co-agonists, the potential of GIP in the treatment of metabolic diseases has been reevaluated. The study of GIP and its co-agonists such as glucagon-like peptide-1 (GLP-1), revealed that its mechanism of action in regulating blood glucose, fat metabolism, and bone metabolism is complex and diverse. A better understanding of GIP evolution can help in designing more effective GIP-based treatment strategies. In this review, we summarize the physiological functions of GIP, systematically explores its diverse structural modifications, delves into the realm of unimolecular multi-agonists, and provides a nuanced portrayal of the developmental prospects of GIP analogs.
Collapse
Affiliation(s)
- Chenxu Zhou
- College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Binbin Gong
- College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Xiyu Liu
- College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Guoqiang Hu
- Taizhou Hospital, Zhejiang University, Taizhou, 317000, China
| | - Lidan Sun
- College of Medicine, Jiaxing University, Jiaxing, 314001, China; Taizhou Hospital, Zhejiang University, Taizhou, 317000, China.
| |
Collapse
|
10
|
Drucker DJ. GLP-1-based therapies for diabetes, obesity and beyond. Nat Rev Drug Discov 2025:10.1038/s41573-025-01183-8. [PMID: 40281304 DOI: 10.1038/s41573-025-01183-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2025] [Indexed: 04/29/2025]
Abstract
Glucagon-like peptide 1 (GLP-1)-based therapies, such as semaglutide and tirzepatide, represent highly effective treatment options for people with type 2 diabetes and obesity, enabling effective control of glucose and weight loss, while reducing cardiovascular and renal morbidity and mortality. The success of these medicines has spurred development of next-generation GLP-1-based drugs, promising greater weight loss, improved tolerability and additional options for the route and frequency of dosing. This Review profiles established and emerging GLP-1-based medicines, discussing optimization of pharmacokinetics and tolerability, engagement of new therapeutically useful pathways and safety aspects. Structurally unique GLP-1-based medicines that achieve substantially greater and rapid weight loss may impact musculoskeletal health, providing a rationale for therapeutics that more selectively target adipose tissue loss while preserving muscle mass and strength. Ongoing clinical trials in peripheral vascular disease, neuropsychiatric and substance use disorders, metabolic liver disease, arthritis, hypertension and neurodegenerative disorders may broaden indications for GLP-1-based therapeutics.
Collapse
Affiliation(s)
- Daniel J Drucker
- Department of Medicine and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Anastasiou IΑ, Argyrakopoulou G, Dalamaga M, Kokkinos A. Dual and Triple Gut Peptide Agonists on the Horizon for the Treatment of Type 2 Diabetes and Obesity. An Overview of Preclinical and Clinical Data. Curr Obes Rep 2025; 14:34. [PMID: 40210807 PMCID: PMC11985575 DOI: 10.1007/s13679-025-00623-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 04/12/2025]
Abstract
PURPOSE OF REVIEW The development of long-acting incretin receptor agonists represents a significant advance in the fight against the concurrent epidemics of type 2 diabetes mellitus (T2DM) and obesity. The aim of the present review is to examine the cellular processes underlying the actions of these new, highly significant classes of peptide receptor agonists. We further explore the potential actions of multi-agonist drugs as well as the mechanisms through which gut-brain communication can be used to achieve long-term weight loss without negative side effects. RECENT FINDINGS Several unimolecular dual-receptor agonists have shown promising clinical efficacy studies when used alone or in conjunction with approved glucose-lowering medications. We also describe the development of incretin-based pharmacotherapy, starting with exendin- 4 and ending with the identification of multi-incretin hormone receptor agonists, which appear to be the next major step in the fight against T2DM and obesity. We discuss the multi-agonists currently in clinical trials and how each new generation of these drugs improves their effectiveness. Since most glucose-dependent insulinotropic polypeptide (GIP) receptor: glucagon-like peptide- 1 receptor (GLP- 1) receptor: glucagon receptor triagonists compete in efficacy with bariatric surgery, the success of these agents in preclinical models and clinical trials suggests a bright future for multi-agonists in the treatment of metabolic diseases. To fully understand how these treatments affect body weight, further research is needed.
Collapse
Affiliation(s)
- Ioanna Α Anastasiou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | | | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Alexander Kokkinos
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| |
Collapse
|
12
|
Zandvakili I, Perez-Tilve D. The unexpected role of GIP in transforming obesity treatment. Trends Endocrinol Metab 2025; 36:330-338. [PMID: 39198118 DOI: 10.1016/j.tem.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Despite sharing incretin activity with glucagon-like peptide 1 (GLP-1), the development of gastric inhibitory polypeptide (GIP)-based drugs has been hindered by the minor effects of native GIP on appetite and body weight and genetic studies associating loss-of-function with reduced obesity. Yet, pharmacologically optimized GIP-based molecules have demonstrated profound weight lowering benefits of GIPR agonism when combined with GLP-1-based therapies, which has re-energized deeper exploration of the molecular mechanisms and downstream signaling of GIPR. Interestingly, both GIPR agonism and antagonism offer metabolic benefits, leading to differing viewpoints on how to target GIPR therapeutically. Here we summarize the emerging evidence about the tissue-specific mechanisms that positions GIP-based therapies as important targets for the next generation of anti-obesity and metabolic therapies.
Collapse
Affiliation(s)
- Inuk Zandvakili
- Division of Digestive Diseases, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Diego Perez-Tilve
- Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
13
|
Al-Zaid B, Al-Sabah S. The C-terminal regions of the GLP-1 and GIP receptors are not the key determinants of their differential arrestin recruitment but modulate the rate of receptor endocytosis. Front Pharmacol 2025; 16:1528295. [PMID: 40201698 PMCID: PMC11975949 DOI: 10.3389/fphar.2025.1528295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/28/2025] [Indexed: 04/10/2025] Open
Abstract
Introduction: Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are important regulators of metabolism and mediate the incretin effect. This glucose-dependent potentiation of insulin secretion is severely impaired in patients with type-2 diabetes mellitus. While pharmacological doses of GLP-1 can overcome this impairment, the same is not true for GIP. The reasons for this are unclear. However, differences in the signalling profiles of the GLP-1 and GIP receptors (GLP-1R and GIPR) may contribute. GLP-1R and GIPR are closely related G protein-coupled receptors but differ in their ability to recruit arrestin, GIPR being relatively poorer. Furthermore, these receptors have been reported to utilize different mechanisms to undergo agonist-induced internalization. Methods: This study aimed to identify the role of the C-terminal region of the two receptors in their differing signalling behaviour using chimeric receptors where the C-terminal tail of one receptor was replaced with that of the other. Results: Replacement of the C-terminal tail had only limited effects on G protein and arrestin recruitment to either receptor. GIP-stimulated internalisation of GIPR occurred at a significantly (P < 0.001) slower rate than GLP-1-stimulated internalisation of GLP-1R. Replacement of the C-terminal tail of GIPR with that of GLP-1R significantly (P < 0.05) increased the internalization rate but not to the rate of wild-type GLP-1R. The reciprocal substitution significantly (P < 0.005) decreased internalization rate. Conclusion: These data show that the C-terminal region of GLP-1R and GIPR is not the critical determinant of their differing ability to recruit arrestin but modulates receptor endocytosis.
Collapse
Affiliation(s)
| | - Suleiman Al-Sabah
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
14
|
James-Okoro PP, Lewis JE, Gribble FM, Reimann F. The role of GIPR in food intake control. Front Endocrinol (Lausanne) 2025; 16:1532076. [PMID: 40166681 PMCID: PMC11955450 DOI: 10.3389/fendo.2025.1532076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is one of two incretin hormones playing key roles in the control of food intake, nutrient assimilation, insulin secretion and whole-body metabolism. Recent pharmacological advances and clinical trials show that unimolecular co-agonists that target the receptors for the incretins - GIP and glucagon-like peptide 1 (GLP-1) - offer more effective treatment strategies for obesity and type 2 diabetes mellitus (T2D) compared with GLP-1 receptor (GLP1R) agonists alone, suggesting previously underappreciated roles of GIP in regulating food intake and body weight. The mechanisms by which GIP regulates energy balance remain controversial as both agonism and antagonism of the GIP receptor (GIPR) produce weight loss and improve metabolic outcomes in preclinical models. Recent studies have shown that GIPR signalling in the central nervous system (CNS), especially in regions of the brain that regulate energy balance, is essential for its action on appetite regulation. This finding has sparked interest in understanding the mechanisms by which GIP engages brain circuits to reduce food intake and body weight. In this review, we present key knowledge around the actions of GIP on food intake regulation and the potential mechanisms by which GIPR and GIPR/GLP1R agonists may regulate energy balance.
Collapse
Affiliation(s)
| | | | - Fiona Mary Gribble
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Frank Reimann
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
15
|
Lorente JS, Sokolov AV, Ferguson G, Schiöth HB, Hauser AS, Gloriam DE. GPCR drug discovery: new agents, targets and indications. Nat Rev Drug Discov 2025:10.1038/s41573-025-01139-y. [PMID: 40033110 DOI: 10.1038/s41573-025-01139-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 03/05/2025]
Abstract
G protein-coupled receptors (GPCRs) form one of the largest drug target families, reflecting their involvement in numerous pathophysiological processes. In this Review, we analyse drug discovery trends for the GPCR superfamily, covering compounds, targets and indications that have reached regulatory approval or that are being investigated in clinical trials. We find that there are 516 approved drugs targeting GPCRs, making up 36% of all approved drugs. These drugs act on 121 GPCR targets, one-third of all non-sensory GPCRs. Furthermore, 337 agents targeting 133 GPCRs, including 30 novel targets, are being investigated in clinical trials. Notably, 165 of these agents are approved drugs being tested for additional indications and novel agents are increasingly allosteric modulators and biologics. Remarkably, diabetes and obesity drugs targeting GPCRs had sales of nearly US $30 billion in 2023 and the numbers of clinical trials for GPCR modulators in the metabolic diseases, oncology and immunology areas are increasing strongly. Finally, we highlight the potential of untapped target-disease associations and pathway-biased signalling. Overall, this Review provides an up-to-date reference for the drugged and potentially druggable GPCRome to inform future GPCR drug discovery and development.
Collapse
Affiliation(s)
- Javier Sánchez Lorente
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aleksandr V Sokolov
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Gavin Ferguson
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- ALPX S.A.S., Grenoble, France
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Alexander S Hauser
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David E Gloriam
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
16
|
Tadross JA, Steuernagel L, Dowsett GKC, Kentistou KA, Lundh S, Porniece M, Klemm P, Rainbow K, Hvid H, Kania K, Polex-Wolf J, Knudsen LB, Pyke C, Perry JRB, Lam BYH, Brüning JC, Yeo GSH. A comprehensive spatio-cellular map of the human hypothalamus. Nature 2025; 639:708-716. [PMID: 39910307 PMCID: PMC11922758 DOI: 10.1038/s41586-024-08504-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/09/2024] [Indexed: 02/07/2025]
Abstract
The hypothalamus is a brain region that plays a key role in coordinating fundamental biological functions1. However, our understanding of the underlying cellular components and neurocircuitries have, until recently, emerged primarily from rodent studies2,3. Here we combine single-nucleus sequencing of 433,369 human hypothalamic cells with spatial transcriptomics, generating a comprehensive spatio-cellular transcriptional map of the hypothalamus, the 'HYPOMAP'. Although conservation of neuronal cell types between humans and mice, as based on transcriptomic identity, is generally high, there are notable exceptions. Specifically, there are significant disparities in the identity of pro-opiomelanocortin neurons and in the expression levels of G-protein-coupled receptors between the two species that carry direct implications for currently approved obesity treatments. Out of the 452 hypothalamic cell types, we find that 291 neuronal clusters are significantly enriched for expression of body mass index (BMI) genome-wide association study genes. This enrichment is driven by 426 'effector' genes. Rare deleterious variants in six of these (MC4R, PCSK1, POMC, CALCR, BSN and CORO1A) associate with BMI at population level, and CORO1A has not been linked previously to BMI. Thus, HYPOMAP provides a detailed atlas of the human hypothalamus in a spatial context and serves as an important resource to identify new druggable targets for treating a wide range of conditions, including reproductive, circadian and metabolic disorders.
Collapse
Affiliation(s)
- John A Tadross
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
- Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Georgina K C Dowsett
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Katherine A Kentistou
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Sofia Lundh
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | - Marta Porniece
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Kara Rainbow
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Henning Hvid
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | - Katarzyna Kania
- Genomics Core, Cancer Research UK Cambridge Institute, Cambridge, UK
| | | | | | - Charles Pyke
- Research & Early Development, Novo Nordisk A/S, Måløv, Denmark
| | - John R B Perry
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Brian Y H Lam
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.
- National Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Giles S H Yeo
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
| |
Collapse
|
17
|
Santi DV. When can weekly anti-obesity peptides be used for monthly administration? Diabetes Obes Metab 2025; 27:1628-1629. [PMID: 39703133 DOI: 10.1111/dom.16134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Affiliation(s)
- Daniel V Santi
- ProLynx, San Francisco, California, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
18
|
Madsbad S, Holst JJ. The promise of glucagon-like peptide 1 receptor agonists (GLP-1RA) for the treatment of obesity: a look at phase 2 and 3 pipelines. Expert Opin Investig Drugs 2025; 34:197-215. [PMID: 40022548 DOI: 10.1080/13543784.2025.2472408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION GLP-1-based therapies have changed the treatment of overweight/obesity. Liraglutide 3.0 mg daily, the first GLP-1 RA approved for treatment of overweight, induced a weight loss of 6-8%, Semaglutide 2.4 mg once weekly improved weight loss to about 12-15%, while the dual GIP/GLP-1 receptor agonist tirzepatide once weekly has induced a weight loss of about 20% in obese people without diabetes. AREAS COVERED This review describes results obtained with GLP-1 mono-agonists, GLP-1/GIP dual agonists, GLP-1/glucagon co-agonists, and the triple agonist retatrutide (GIP/GLP-1/glucagon), which have shown beneficial effect both on body weight and steatotic liver disease. A combination of semaglutide (a GLP-1 agonist) and cagrilintide (a long-acting amylin analogue) for weekly administration is currently in phase III development, and so is oral semaglutide and several non-peptide small molecule GLP-1 agonists for oral administration. The adverse events with the GLP-1-based therapies are primarily gastrointestinal and include nausea, vomiting, obstipation, or diarrhea, which often can be mitigated by slow up titration. EXPERT OPINION The GLP-1-based therapies will change the treatment of obesity and its comorbidities including steatotic liver disease in the future. Outstanding question is maintenance of the weight loss, possibly pharmacological treatment needs to be life-long.
Collapse
Affiliation(s)
- Sten Madsbad
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- The NovoNordisk Foundation Centre for Basic Metabolic Research and the Department of Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Schneider EL, Hangasky JA, Ashley GW, Santi DV. Long-Acting, Longer-Acting, and Ultralong-Acting Antiobesity Peptides. J Med Chem 2025; 68:2301-2306. [PMID: 39895561 DOI: 10.1021/acs.jmedchem.4c02647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
This perspective describes the current status and future prospects of developing long- to ultralong-acting anti-obesity peptides. First, we discuss the current status of lipidation, PEGylation, and Fc fusion technologies to obtain long-acting peptides administered once weekly, and we critique their proposed use for longer dosing intervals. Next, we describe the approach and current results of using macromolecular peptide prodrugs with preprogrammed releasable linkers to achieve longer-acting peptides that can be administered weekly or monthly. Finally, we posit novel modifications of the latter technology that could provide ultralong half-lives of over one month by advantageously exploiting the clearance rates of released peptides. As examples, we posit that prodrugs of low-clearance GLP-1 agonists derived from peptides already modified by lipidation, PEGylation, and Fc fusion could produce ultralong-acting agonists with dosing intervals reaching 3 to even 6 months.
Collapse
Affiliation(s)
- Eric L Schneider
- ProLynx, Inc., 135 Mississippi Street, San Francisco, California 94107, United States
| | - John A Hangasky
- ProLynx, Inc., 135 Mississippi Street, San Francisco, California 94107, United States
| | - Gary W Ashley
- ProLynx, Inc., 135 Mississippi Street, San Francisco, California 94107, United States
| | - Daniel V Santi
- ProLynx, Inc., 135 Mississippi Street, San Francisco, California 94107, United States
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| |
Collapse
|
20
|
Fan S, Li J, Zhuang J, Zhou Q, Mai Y, Lin B, Wang MW, Wu C. Disulfide-Directed Multicyclic Peptides with N-Terminally Extendable α-Helices for Recognition and Activation of G Protein-Coupled Receptors. J Am Chem Soc 2025; 147:4821-4832. [PMID: 39688263 DOI: 10.1021/jacs.4c12808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Many peptide hormones adopt long α-helical structures upon interacting with their cognate receptors but often exhibit flexible conformations when unbound. Strategies that can stabilize long α-helices without disrupting their binding to receptors are still lacking, which hinders progress in their biological applications and drug development. Here, we present an approach that combines rational design with library screening to create and identify a unique disulfide-directed multicyclic peptide (DDMP) scaffold, which could effectively stabilize N-terminally extendable α-helices while displaying exceptional efficiency in disulfide pairing and oxidative folding. This DDMP scaffold was then utilized for stabilizing the α-helical structure of glucagon-like peptide-1 (GLP-1), resulting in a potent GLP-1 receptor (GLP-1R) agonist with a significantly improved α-helicity and proteolytic stability. By incorporating external α-helices into the DDMP scaffold, we can effectively preserve the native N-terminal α-helical structures while allowing for extensive evolution of the C-terminal disulfide-rich domain for enhancing target binding, as demonstrated by the generation of the DDMP-stabilized GLP-1 (g1:Ox). The cryo-electron microscopy structure of the g1:Ox-GLP-1R in complex with heterotrimeric Gs reveals the molecular basis for the potent binding between g1:Ox and GLP-1R. Specifically, the DDMP moiety establishes additional interactions with the extracellular domain of GLP-1R, which are absent in the case of GLP-1. Thus, this work offers a novel and effective approach for engineering therapeutic peptides and other peptide α-helices, ensuring that both the N- and C-terminal regions remain essential for target recognition and activation.
Collapse
Affiliation(s)
- Shihui Fan
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jie Li
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jie Zhuang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Qingtong Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Research Center for Deepsea Bioresources, Sanya, Hainan 572025, China
| | - Yiting Mai
- Research Center for Deepsea Bioresources, Sanya, Hainan 572025, China
| | - Bingni Lin
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Ming-Wei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Research Center for Deepsea Bioresources, Sanya, Hainan 572025, China
- Research Center for Medicinal Structural Biology, National Research Center for Translational Medicine at Shanghai, State Key Laboratory of Medical Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, School of Pharmacy, Hainan Medical University, Haikou 570228, China
| | - Chuanliu Wu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
21
|
Dash S. Obesity and Cardiometabolic Disease: Insights From Genetic Studies. Can J Cardiol 2025:S0828-282X(25)00104-7. [PMID: 39920990 DOI: 10.1016/j.cjca.2025.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/10/2025] Open
Abstract
Obesity is a highly prevalent chronic disease and major driver of both atherosclerotic heart disease and heart failure. Obesity is also a heritable neuronal disease with heritability estimates of up to 70%. In this work I review how common genetic variants, usually with small effect sizes, contribute to the risk for developing obesity and cardiometabolic disease in the majority of people and how this can be further modulated by environmental factors. In some individuals, rare genetic variants with large effect sizes can influence the risk of developing obesity, in some cases in a Mendelian manner. I also address how identification of these rare variants has led to fundamental biologic insights into how satiety and reward are biologic processes, has led to more personalized treatments, and has identified potential novel drug treatments. Biologic insights derived from genetic studies of obesity have also improved our understanding of the causal mediators between obesity and cardiovascular disease. A major limitation of studies to date is that they involved mostly people of European ancestry. Studying more diverse populations will improve our understanding of obesity and cardiometabolic disease. Lessons derived from genetic studies make a compelling case for increasing accessibility to therapies that have sustained efficacy in managing obesity and improving health. This increased knowledge must also inform public health initiatives that will reduce the prevalence of obesity in the coming decades.
Collapse
Affiliation(s)
- Satya Dash
- Department of Medicine, University of Toronto and University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
22
|
Douros JD, Flak JN, Knerr PJ. The agony and the efficacy: central mechanisms of GLP-1 induced adverse events and their mitigation by GIP. Front Endocrinol (Lausanne) 2025; 16:1530985. [PMID: 39963285 PMCID: PMC11830610 DOI: 10.3389/fendo.2025.1530985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
| | - Jonathan N. Flak
- Indiana Biosciences Research Institute, Indianapolis, IN, United States
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Patrick J. Knerr
- Indiana Biosciences Research Institute, Indianapolis, IN, United States
| |
Collapse
|
23
|
Li C, Yuan Y, Jia Y, Zhou Q, Wang Q, Jiang X. Cellular senescence: from homeostasis to pathological implications and therapeutic strategies. Front Immunol 2025; 16:1534263. [PMID: 39963130 PMCID: PMC11830604 DOI: 10.3389/fimmu.2025.1534263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Cellular aging is a multifactorial and intricately regulated physiological process with profound implications. The interaction between cellular senescence and cancer is complex and multifaceted, senescence can both promote and inhibit tumor progression through various mechanisms. M6A methylation modification regulates the aging process of cells and tissues by modulating senescence-related genes. In this review, we comprehensively discuss the characteristics of cellular senescence, the signaling pathways regulating senescence, the biomarkers of senescence, and the mechanisms of anti-senescence drugs. Notably, this review also delves into the complex interactions between senescence and cancer, emphasizing the dual role of the senescent microenvironment in tumor initiation, progression, and treatment. Finally, we thoroughly explore the function and mechanism of m6A methylation modification in cellular senescence, revealing its critical role in regulating gene expression and maintaining cellular homeostasis. In conclusion, this review provides a comprehensive perspective on the molecular mechanisms and biological significance of cellular senescence and offers new insights for the development of anti-senescence strategies.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Yixiao Yuan
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| | - YingDong Jia
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Zhou
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Xiulin Jiang
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|
24
|
Davies I, Adriaenssens AE, Scott WR, Carling D, Murphy KG, Minnion JS, Bloom SR, Jones B, Tan TMM. Chronic GIPR agonism results in pancreatic islet GIPR functional desensitisation. Mol Metab 2025; 92:102094. [PMID: 39788289 PMCID: PMC11786100 DOI: 10.1016/j.molmet.2025.102094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVES There is renewed interest in targeting the glucose-dependent insulinotropic polypeptide receptor (GIPR) for treatment of obesity and type 2 diabetes. G-protein coupled receptor desensitisation is suggested to reduce the long-term efficacy of glucagon-like-peptide 1 receptor (GLP-1R) agonists and may similarly affect the efficacy of GIPR agonists. We explored the extent of pancreatic GIPR functional desensitisation with sustained agonist exposure. METHODS A long-acting GIPR agonist, GIP108, was used to probe the effect of sustained agonist exposure on cAMP responses in dispersed pancreatic islets using live cell imaging, with rechallenge cAMP responses after prior agonist treatment used to quantify functional desensitisation. Receptor internalisation and β-arrestin-2 activation were investigated in vitro using imaging-based assays. Pancreatic mouse GIPR desensitisation was assessed in vivo via intraperitoneal glucose tolerance testing. RESULTS GIP108 treatment led to weight loss and improved glucose homeostasis in mice. Prolonged exposure to GIPR agonists produced homologous functional GIPR desensitisation in isolated islets. GIP108 pre-treatment in vivo also reduced the subsequent anti-hyperglycaemic response to GIP re-challenge. GIPR showed minimal agonist-induced internalisation or β-arrestin-2 activation. CONCLUSIONS Although GIP108 chronic treatment improved glucose tolerance, it also resulted in partial desensitisation of the pancreatic islet GIPR. This suggests that ligands with reduced desensitisation tendency might lead to improved in vivo efficacy. Understanding whether pancreatic GIPR desensitisation affects the long-term benefits of GIPR agonists in humans is vital to design effective metabolic pharmacotherapies.
Collapse
Affiliation(s)
- Iona Davies
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom
| | - Alice E Adriaenssens
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, United Kingdom
| | - William R Scott
- MRC Laboratory of Medical Sciences, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - David Carling
- MRC Laboratory of Medical Sciences, London, United Kingdom
| | - Kevin G Murphy
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom
| | - James S Minnion
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom
| | - Stephen R Bloom
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom
| | - Ben Jones
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom.
| | - Tricia M-M Tan
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom.
| |
Collapse
|
25
|
Manchanda Y, Jones B, Carrat G, Ramchunder Z, Marchetti P, Leclerc I, Thennati R, Burade V, Natarajan M, Shahi P, Tomas A, Rutter GA. Binding Kinetics, Bias, Receptor Internalization and Effects on Insulin Secretion in vitro and in vivo of a Novel GLP-1R/GIPR Dual Agonist, HISHS-2001. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632834. [PMID: 39868265 PMCID: PMC11760779 DOI: 10.1101/2025.01.13.632834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The use of incretin analogues has emerged in recent years as an effective approach to achieve both enhanced insulin secretion and weight loss in type 2 diabetes (T2D) patients. Agonists which bind and stimulate multiple receptors have shown particular promise. However, off target effects, including nausea and diarrhoea, remain a complication of using these agents, and modified versions with optimized pharmacological profiles and/or biased signaling at the cognate receptors are increasingly sought. Here, we describe the synthesis and properties of a molecule which binds to both glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors (GLP-1R and GIPR) to enhance insulin secretion. HISHS-2001 shows increased affinity at the GLP-1R, as well as a tendency towards reduced internalization and recycling at this receptor versus FDA-approved dual GLP-1R/GIPR agonist tirzepatide. HISHS-2001 also displayed significantly greater bias towards cAMP generation versus β-arrestin 2 recruitment compared to tirzepatide. In contrast, Gαs recruitment was lower versus tirzepatide at the GLP-1R, but higher at the GIPR. Administered to obese hyperglycaemic db/db mice, HISHS-2001 increased circulating insulin whilst lowering body weight and HbA1c with similar efficacy to tirzepatide at substantially lower doses. Thus, HISHS-2001 represents a novel dual receptor agonist with an improved pharmacological profile.
Collapse
Affiliation(s)
- Yusman Manchanda
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ben Jones
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Gaelle Carrat
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Zenouska Ramchunder
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Piero Marchetti
- Department of Experimental and Clinical Medicine, Pancreatic Islets Laboratory, University of Pisa, Pisa, Italy
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
- Centre de Recherche du Centre Hôspitalier de l’Université de Montreal (CRCHUM) et Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | | | - Vinod Burade
- Sun Pharmaceutical Industries Limited, Vadodara, Gujarat, India
| | | | - Pradeep Shahi
- Sun Pharmaceutical Industries Limited, Vadodara, Gujarat, India
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
- Sun Pharmaceutical Industries Limited, Vadodara, Gujarat, India
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Research Institute of the McGill University Health Sciences Centre, Montreal, QC, Canada
| |
Collapse
|
26
|
ÇETİN E, PEDERSEN B, BURAK MF. Paradigm shift in obesity treatment: an extensive review of current pipeline agents. Turk J Med Sci 2025; 55:1-16. [PMID: 40104296 PMCID: PMC11913498 DOI: 10.55730/1300-0144.5938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/18/2025] [Accepted: 01/15/2025] [Indexed: 03/20/2025] Open
Abstract
Obesity is a multifaceted disease that poses a significant public health challenge. Recent discoveries in understanding the biological pathways that regulate satiety and metabolism have led to a shift in the treatment paradigm for obesity. Thus, the gap between pharmacological and surgical interventions has diminished. The latest approved antiobesity medications help to achieve weight loss comparable to surgery. These GLP-1 analog-based therapies not only cause substantial weight loss but also improve obesity-associated comorbidities. However, there are still unmet needs in obesity care, and treatment options with alternative pathways are necessary. Whether achieved through lifestyle changes or medication, weight loss often leads to muscle mass loss and reduced energy expenditure, resulting in rebound weight gain. Moreover, addressing severe obesity and comorbidities, such as metabolic-associated fatty liver disease (MAFLD), metabolic dysfunction-associated steatohepatitis (MASH), heart failure with preserved ejection fraction, and obstructive sleep apnea, necessitates the development of additional therapeutic strategies. Various antiobesity medications with novel mechanisms of action are currently in the pipeline. Myostatin-activin pathway inhibitors are under development to preserve muscle mass, and combination therapies with glucagon agonists address MAFLD and MASH. Amylin agonists offer a promising alternative to those unable to tolerate GLP-1 analogs. Mitochondrial uncouplers are under investigation for enhancing energy expenditure, NLRP-3 inhibitors for reducing inflammation, and GWAS targets for additional weight loss benefits. Combination therapies, such as dual or triple hormonal receptor agonists, are being developed to maximize weight loss and optimize tolerability. These emerging medications in the clinical trial pipeline show promise for more tolerable and sustainable obesity management.
Collapse
Affiliation(s)
- Ecesu ÇETİN
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,
United States
| | - Brian PEDERSEN
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,
United States
| | - Mehmet Furkan BURAK
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA,
United States
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA,
United States
| |
Collapse
|
27
|
Jones LA, Brierley DI. GLP-1 and the Neurobiology of Eating Control: Recent Advances. Endocrinology 2025; 166:bqae167. [PMID: 39813121 PMCID: PMC11745901 DOI: 10.1210/endocr/bqae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/09/2024] [Accepted: 12/31/2024] [Indexed: 01/18/2025]
Abstract
Obesity is now considered a chronic relapsing progressive disease, associated with increased all-cause mortality that scales with body weight, affecting more than 1 billion people worldwide. Excess body fat is strongly associated with excess energy intake, and most successful anti-obesity medications (AOMs) counter this positive energy balance through the suppression of eating to drive weight loss. Historically, AOMs have been characterized by modest weight loss and side effects which are compliance-limiting, and in some cases life-threatening. However, the field of obesity pharmacotherapy has now entered a new era of AOMs based on analogues of the gut hormone and neuropeptide glucagon-like peptide-1 (GLP-1). The latest versions of these drugs elicit unprecedented levels of weight loss in clinical trials, which are now starting to be substantiated in real-world usage. Notably, these drugs reduce weight primarily by reducing energy intake, via activation of the GLP-1 receptor on multiple sites of action primarily in the central nervous system, although the most relevant sites of action, and the neural circuits recruited remain contentious. Here we provide a targeted synthesis of recent developments in the field of GLP-1 neurobiology, highlighting studies which have advanced our understanding of how GLP-1 signaling modulates eating, and identify open questions and future challenges we believe still need to be addressed to aid the prevention and/or treatment of obesity.
Collapse
Affiliation(s)
- Lauren A Jones
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6DE, UK
| | - Daniel I Brierley
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6DE, UK
| |
Collapse
|
28
|
Kim MK, Kim HS. An Overview of Existing and Emerging Weight-Loss Drugs to Target Obesity-Related Complications: Insights from Clinical Trials. Biomol Ther (Seoul) 2025; 33:5-17. [PMID: 39696983 PMCID: PMC11704407 DOI: 10.4062/biomolther.2024.228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
Obesity requires treatment as it is associated with health problems such as type 2 diabetes, hypertension, dyslipidemia, cardiovascular diseases, and some cancers, which increase mortality rates. Achieving sufficient weight loss to reduce obesity-related diseases requires a variety of interventions, including comprehensive lifestyle modification of diet and exercise, change in behavior, anti-obesity medications, and surgery. To date, anti-obesity agents with various mechanisms of action have been developed, and mostly reduce energy intake, resulting in weight loss of about 5% to 10% compared to baseline. Recently developed drugs and those currently under development have been shown to reduce body weight by more than 10% and are expected to reduce obesity-related complications. This article summarizes existing and emerging anti-obesity medications, with a particular focus on those evaluated in clinical trials.
Collapse
Affiliation(s)
- Mi Kyung Kim
- Department of Internal Medicine, Keimyung University, School of Medicine, Dongsan Hospital, Daegu 42601, Republic of Korea
- Center of Bariatric and Metabolic Surgery, Keimyung University, Dongsan Hospital, Daegu 42601, Republic of Korea
| | - Hye Soon Kim
- Department of Internal Medicine, Keimyung University, School of Medicine, Dongsan Hospital, Daegu 42601, Republic of Korea
- Center of Bariatric and Metabolic Surgery, Keimyung University, Dongsan Hospital, Daegu 42601, Republic of Korea
| |
Collapse
|
29
|
Kokkorakis M, Chakhtoura M, Rhayem C, Al Rifai J, Ghezzawi M, Valenzuela-Vallejo L, Mantzoros CS. Emerging pharmacotherapies for obesity: A systematic review. Pharmacol Rev 2025; 77:100002. [PMID: 39952695 DOI: 10.1124/pharmrev.123.001045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
The history of antiobesity pharmacotherapies is marked by disappointments, often entangled with societal pressure promoting weight loss and the prevailing conviction that excess body weight signifies a lack of willpower. However, categories of emerging pharmacotherapies generate hope to reduce obesity rates. This systematic review of phase 2 and phase 3 trials in adults with overweight/obesity investigates the effect of novel weight loss pharmacotherapies, compared to placebo/control or US Food and Drug Administration-approved weight loss medication, through searching Medline, Embase, and ClinicalTrials.gov (2012-2024). We identified 53 phase 3 and phase 2 trials, with 36 emerging antiobesity drugs or combinations thereof and 4 withdrawn or terminated trials. Oral semaglutide 50 mg is the only medication that has completed a phase 3 trial. There are 14 ongoing phase 3 trials on glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) (ecnoglutide, orforglipron, and TG103), GLP-1 RA/amylin agonist (CagriSema), GLP-1/glucagon RAs (mazdutide and survodutide), GLP-1/glucose-dependent insulinotropic polypeptide and glucagon RA (retatrutide), dapagliflozin, and the combination sibutramine/topiramate. Completed phase 2 trials on incretin-based therapies showed a mean percent weight loss of 7.4% to 24.2%. Almost half of the drugs undergoing phase 2 trials are incretin analogs. The obesity drug pipeline is expanding rapidly, with the most promising results reported with incretin analogs. Data on mortality and obesity-related complications, such as cardio-renal-metabolic events, are needed. Moreover, long-term follow-up data on the safety and efficacy of weight maintenance with novel obesity pharmacotherapies, along with studies focused on underrepresented populations, cost-effectiveness assessments, and drug availability, are needed to bridge the care gap for patients with obesity. SIGNIFICANCE STATEMENT: Obesity is the epidemic of the 21st century. Except for the newer injectable medications, drugs with suboptimal efficacy have been available in the clinician's armamentarium for weight management. However, emerging alternatives of novel agents and combinations populate the current obesity therapeutic pipeline. This systematic review identifies the state and mechanism of action of emerging pharmacotherapies undergoing or having completed phase 2 and phase 3 clinical trials. The information provided herein furthers the understanding of obesity management, implying direct clinical implications and stimulating research initiatives.
Collapse
Affiliation(s)
- Michail Kokkorakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marlene Chakhtoura
- Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| | - Caline Rhayem
- Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jana Al Rifai
- Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Malak Ghezzawi
- Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Laura Valenzuela-Vallejo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
30
|
Hvisdas CM, Goode ND, Kim DH, Silvey MJ, Flood JJ. Characterization of Interchanging Incretin Analogs in Clinical Practice: A Descriptive Report. Endocr Pract 2025; 31:59-64. [PMID: 39414234 DOI: 10.1016/j.eprac.2024.09.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024]
Abstract
OBJECTIVE To characterize the tolerability associated with incretin analog interchanges to equipotent or higher strengths based on an interchange process in an adult outpatient setting. METHODS This was a retrospective chart review of adult patients receiving care through a participating family medicine or endocrinology clinic between January 1, 2022, and November 30, 2022 at a major academic medical center. An incretin analog equivalency table and protocol for interchange was created in response to ongoing shortages and need for therapy adjustments to different medications within the same class. Patients were included if a recommended incretin analog interchange was initiated and a tolerability assessment was conducted. Patients were excluded if they did not meet inclusion criteria or if they were unreachable for tolerability assessments for interchanged agents. RESULTS There were 156 patients included for characterization and response to tolerability of interchange. It was determined that 96% of patients tolerated the incretin analog interchange. A dose escalation occurred in 58% of patients, 41% were transitioned to an equipotent dose, and a dose decrease was considered in 1 patient. Prior authorizations were required 74% of the time for the new therapy. The most common interchanges were dulaglutide 4.5 mg to tirzepatide 7.5 mg, dulaglutide 4.5 mg to tirzepatide 10 mg, and dulaglutide 3 mg to tirzepatide 7.5 mg. These interchanges made up 37.3% of the total population and were observed to have 93% tolerability. Seven patients did not tolerate incretin analog interchange. Five experienced gastrointestinal effects and 2 experienced injection site reactions. The interchange of incretin analog was estimated to reduce time to maximum dose by a median of 3 months. During this study, no patients experienced interruption of therapy defined as missing a dose of incretin analog. CONCLUSION This characterization report demonstrates an effective approach to addressing incretin analog interchanges. A high level of tolerability is evident with the defined interchange process. Future studies should continue to confirm effective and safe interchanges of incretin analogs from outcomes and tolerability reports.
Collapse
Affiliation(s)
- Christopher M Hvisdas
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania.
| | - Natalie D Goode
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania
| | - Diane H Kim
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania
| | - Michael J Silvey
- Department of Pharmacy, Penn Presbyterian Medical Center, University of Pennsylvania Health System, Philadelphia, Pennsylvania
| | - Jeremy J Flood
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Hamza M, Papamargaritis D, Davies MJ. Tirzepatide for overweight and obesity management. Expert Opin Pharmacother 2025; 26:31-49. [PMID: 39632534 DOI: 10.1080/14656566.2024.2436595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Tirzepatide is a once-weekly dual agonist, acting on glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors. It is approved at the same doses (5, 10 and 15 mg) for both type 2 diabetes (T2D) and chronic weight management. AREAS COVERED Following a search in PubMed, clinicaltrials.gov, conference abstracts and Lilly website, we review herein the global phase 3 SURMOUNT program on tirzepatide's safety and efficacy for chronic weight management. Additionally, we discuss findings from the regional SURMOUNT-CN and SURMOUNT-J trials (in East-Asian populations) and the phase 2 SYNERGY-NASH, phase 3 SURMOUNT-OSA and SUMMIT studies on tirzepatide's impact on obesity-related complications. We also explore the clinical implications of SURMOUNT program results, considerations for tirzepatide prescribing for overweight/obesity, ongoing research and evidence gaps. EXPERT OPINION Tirzepatide marks a new era in overweight/obesity treatment, enabling many to achieve ≥ 20% weight loss. It is well-tolerated with a safety profile similar to GLP-1 receptor agonists. Tirzepatide also results in clinically important improvements in multiple obesity-related complications including sleep apnea, metabolic-dysfunction associated steatohepatitis, heart failure with preserved ejection fraction and diabetes prevention. Ongoing trials will provide further data on tirzepatide's long-term safety, efficacy (including cardiovascular outcomes) and potential cost-effectiveness for managing overweight/obesity and/or T2D.
Collapse
Affiliation(s)
- Malak Hamza
- Diabetes Research Centre, University of Leicester, Leicester, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Dimitris Papamargaritis
- Diabetes Research Centre, University of Leicester, Leicester, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester, UK
- Department of Diabetes and Endocrinology, Kettering General Hospital NHS Trust, Kettering, UK
| | - Melanie J Davies
- Diabetes Research Centre, University of Leicester, Leicester, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
32
|
Zhou Q, Li G, Hang K, Li J, Yang D, Wang MW. Weight Loss Blockbuster Development: A Role for Unimolecular Polypharmacology. Annu Rev Pharmacol Toxicol 2025; 65:191-213. [PMID: 39259982 DOI: 10.1146/annurev-pharmtox-061324-011832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) impact more than 2.5 billion adults worldwide, necessitating innovative therapeutic approaches. Unimolecular polypharmacology, which involves designing single molecules to target multiple receptors or pathways simultaneously, has revolutionized treatment strategies. Blockbuster drugs such as tirzepatide and retatrutide have shown unprecedented success in managing obesity and T2DM, demonstrating superior efficacy compared to conventional single agonists. Tirzepatide, in particular, has garnered tremendous attention for its remarkable effectiveness in promoting weight loss and improving glycemic control, while offering additional cardiovascular and renal benefits. Despite their promises, such therapeutic agents also face challenges that include gastrointestinal side effects, patient compliance issues, and body weight rebound after cessation of the treatment. Nonetheless, the development of these therapies marks a significant leap forward, underscoring the transformative potential of unimolecular polypharmacology in addressing metabolic diseases and paving the way for future innovations in personalized medicine.
Collapse
Affiliation(s)
- Qingtong Zhou
- Research Center for Deepsea Bioresources, Sanya, Hainan, China
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Guanyi Li
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
- Research Center for Deepsea Bioresources, Sanya, Hainan, China
| | - Kaini Hang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jie Li
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dehua Yang
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Chemical Biology and The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China;
- Research Center for Deepsea Bioresources, Sanya, Hainan, China
| | - Ming-Wei Wang
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, School of Pharmacy, Hainan Medical University, Haikou, China
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan
- State Key Laboratory of Chemical Biology and The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China;
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Research Center for Deepsea Bioresources, Sanya, Hainan, China
- Translational Research Center for Structural Biology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Koohi F, Harshfield EL, Gill D, Ge W, Burgess S, Markus HS. Optimizing treatment of cardiovascular risk factors in cerebral small vessel disease using genetics. Brain 2024:awae399. [PMID: 39661645 PMCID: PMC7617411 DOI: 10.1093/brain/awae399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/18/2024] [Accepted: 12/10/2024] [Indexed: 12/13/2024] Open
Abstract
Cerebral small vessel disease (cSVD) causes lacunar stroke (LS), intracerebral haemorrhage, and is the most common pathology underlying vascular dementia. However, there are few trials examining whether treating conventional cardiovascular risk factors reduce stroke risk in cSVD, as opposed to stroke as a whole. We used Mendelian randomization techniques to investigate which risk factors are causally related to cSVD and to evaluate whether specific drugs may be beneficial in cSVD prevention. We identified genetic proxies for blood pressure traits, lipids, glycaemic markers, anthropometry measures, smoking, alcohol consumption, and physical activity from large-scale genome-wide association studies of European ancestry. We also selected genetic variants as proxies for drug target perturbation in hypertension, dyslipidaemia, hyperglycaemia, and obesity. Mendelian randomization was performed to assess their associations with LS from the GIGASTROKE Consortium (n = 6811) and in a sensitivity analysis in a cohort of patients with MRI-confirmed LS (n = 3306). We also investigated associations with three neuroimaging features of cSVD, namely, white matter hyperintensities (n = 55 291), fractional anisotropy (n = 36 460), and mean diffusivity (n = 36 012). Genetic predisposition to higher systolic and diastolic blood pressure was associated with LS and cSVD imaging markers. Genetically predicted liability to diabetes, obesity, smoking, higher triglyceride levels, and the ratio of triglycerides to high density lipoprotein (HDL) also showed detrimental associations with LS risk, while genetic predisposition to higher HDL concentrations and moderate-to-vigorous physical activity showed protective associations. Genetically proxied blood pressure-lowering through calcium channel blockers (CCBs) was associated with cSVD imaging markers, while genetically proxied HDL-raising through Cholesteryl Ester Transfer Protein (CETP) inhibitors, triglyceride-lowering through lipoprotein lipase (LPL), and weight-lowering through gastric inhibitory polypeptide receptor (GIPR) were associated with lower risk of LS. Our findings highlight the importance of some conventional cardiovascular risk factors, including blood pressure and BMI, in cSVD, but not other e.g. LDL. The findings further demonstrate the potential beneficial effects of CCBs on cSVD imaging markers and CETP inhibitors, LPL enhancement, and GIPR obesity-targeted drugs on LS. They provide useful information for initiating future clinical trials examining secondary prevention strategies in cSVD.
Collapse
Affiliation(s)
- Fatemeh Koohi
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Eric L Harshfield
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, W12 0BZ, UK
| | - Wenjing Ge
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, CB2 0SR, UK
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| |
Collapse
|
34
|
Abasheva D, Ortiz A, Fernandez-Fernandez B. GLP-1 receptor agonists in patients with chronic kidney disease and either overweight or obesity. Clin Kidney J 2024; 17:19-35. [PMID: 39583142 PMCID: PMC11581768 DOI: 10.1093/ckj/sfae296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Indexed: 11/26/2024] Open
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) have emerged as game-changers across the cardiovascular-kidney-metabolic (CKM) spectrum: overweight/obesity, type 2 diabetes mellitus (T2DM) and associated chronic kidney disease (CKD) and cardiovascular disease (CVD). Liraglutide, semaglutide and tirzepatide are European Medicines Agency approved to improve metabolic control in T2DM and to decrease weight in persons with obesity [body mass index (BMI) ≥30 kg/m2] or with overweight (BMI ≥27 kg/m2) associated with weight-related comorbidities such as hypertension, dyslipidaemia, CVD and others. Additionally, liraglutide and semaglutide are approved to reduce CVD risk in patients with CVD and T2DM. Semaglutide is also approved to reduce CVD risk in patients with CVD and either obesity or overweight and in phase 3 clinical trials showed kidney and cardiovascular protection in patients with T2DM and albuminuric CKD (FLOW trial) as well as in persons without diabetes that had CVD and overweight/obesity (SELECT trial). Thus, nephrologists should consider prescribing GLP-1 RAs to improve metabolic control, reduce CVD risk or improve kidney outcomes in three scenarios: patients with overweight and a related comorbid condition such as hypertension, dyslipidaemia or CVD, patients with obesity and patients with T2DM. This review addresses the promising landscape of GLP-1 RAs to treat persons with overweight or obesity, with or without T2DM, within the context of CKD, assessing their safety and impact on weight, metabolic control, blood pressure and kidney and cardiovascular outcomes, as part of a holistic patient-centred approach to preserve CKM health.
Collapse
Affiliation(s)
- Daria Abasheva
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Beatriz Fernandez-Fernandez
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
35
|
Messineo L, Bakker JP, Cronin J, Yee J, White DP. Obstructive sleep apnea and obesity: A review of epidemiology, pathophysiology and the effect of weight-loss treatments. Sleep Med Rev 2024; 78:101996. [PMID: 39244884 DOI: 10.1016/j.smrv.2024.101996] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/13/2024] [Accepted: 08/23/2024] [Indexed: 09/10/2024]
Abstract
Despite the commonly-accepted paradigm that patients with obstructive sleep apnea (OSA) also invariably have obesity, OSA prevalence extends beyond obesity. This necessitates a reevaluation of screening strategies, biomarkers of increased OSA risk, and heightened awareness among healthcare providers about the array of OSA treatments for diverse adult populations. While obesity contributes importantly to OSA pathogenesis, there is substantial evidence that non-anatomical factors also play a crucial role, especially in patients who do not have obesity. In recent years, notwithstanding the recognition of diverse contributors to OSA pathogenesis, research has frequently focused on weight reduction to address OSA. Insights from past experiences with bariatric surgery in OSA serve as a lens to anticipate potential outcomes of emerging anti-obesity pharmacotherapies. Pharmacological alternatives, particularly incretin agonists, exhibit promise in weight reduction and OSA improvement, but encounter obstacles such as potential side effects and high costs. With this comprehensive narrative review, we delve into the complex epidemiological and pathophysiological connections between OSA and obesity. Additionally, we emphasize the importance of a multifaceted approach to OSA treatment, recognizing that while weight management is crucial, there is a need for comprehensive strategies that go beyond traditional weight-centric perspectives.
Collapse
Affiliation(s)
- Ludovico Messineo
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, USA.
| | - Jessie P Bakker
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, USA
| | | | | | - David P White
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
Novikoff A, Grandl G, Liu X, D. Müller T. Why are we still in need for novel anti-obesity medications? THE LANCET REGIONAL HEALTH. EUROPE 2024; 47:101098. [PMID: 39726721 PMCID: PMC11670685 DOI: 10.1016/j.lanepe.2024.101098] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 12/28/2024]
Abstract
From the pioneering moment in 1987 when the insulinotropic effect of glucagon-like peptide 1 (GLP-1) was first demonstrated in humans, to today's pharmaceutical gold rush for GLP-1-based treatments of obesity, the journey of GLP-1 pharmacology has been nothing short of extraordinary. The sequential conceptual developments of long-acting GLP-1 receptor (GLP-1R) mono-agonists, GLP-1R/glucose-dependent insulinotropic polypeptide receptor (GIPR) dual-agonists, and GLP-1R/GIPR/glucagon receptor (GcgR) triple agonists, have led to profound body weight-lowering capacities, with benefits that extend past obesity and towards obesity-associated diseases. The GLP-1R/GIPR dual-agonist tirzepatide has demonstrated a remarkable 23% body weight reduction in individuals with obesity over 72 weeks, eclipsing the average result achieved by certain types of bariatric surgery. Meanwhile, the GLP-1R/GIPR/GcgR triple-agonist retatrutide achieves similar body weight loss (∼25%) in just two-thirds of the time, potentially surpassing the efficacy of Roux-en-Y gastric bypass. These remarkable achievements rightfully raise the question whether and why there is still need for novel anti-obesity medications (AOMs) in the future.
Collapse
Affiliation(s)
- Aaron Novikoff
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Gerald Grandl
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Xue Liu
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Timo D. Müller
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Germany
| |
Collapse
|
37
|
Grandl G, Novikoff A, Liu X, Müller TD. Recent achievements and future directions of anti-obesity medications. THE LANCET REGIONAL HEALTH. EUROPE 2024; 47:101100. [PMID: 39582489 PMCID: PMC11585837 DOI: 10.1016/j.lanepe.2024.101100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/06/2024] [Accepted: 10/03/2024] [Indexed: 11/26/2024]
Abstract
Pharmacological management of obesity long suffered from a reputation of a 'Mission Impossible,' with inefficient weight loss and/or unacceptable tolerability. However, the tide has turned with recent progress in biochemical engineering and the development of long-acting agonists at the receptor for glucagon-like peptide-1 (GLP-1), and with unimolecular peptides that simultaneously possess activity at the receptors for GLP-1, the glucose-dependent insulinotropic polypeptide (GIP) and glucagon. Some of these novel therapeutics not only improve body weight and glycemic control in individuals with obesity and type 2 diabetes with hitherto unmet efficacy and tolerable safety, but also exhibit potential therapeutic value in diverse areas such as neurodegenerative diseases, fatty liver disease, dyslipidemia, atherosclerosis, and cardiovascular diseases. In this review, we highlight recent advances in incretin-based therapies and discuss their pharmacological potential within and beyond the treatment of obesity and diabetes, as well as their limitations in use, side effects, and underlying molecular mechanisms.
Collapse
Affiliation(s)
- Gerald Grandl
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Aaron Novikoff
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Xue Liu
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Timo D. Müller
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Germany
| |
Collapse
|
38
|
Sun X, Shukla M, Wang W, Li S. Unlocking gut-liver-brain axis communication metabolites: energy metabolism, immunity and barriers. NPJ Biofilms Microbiomes 2024; 10:136. [PMID: 39587086 PMCID: PMC11589602 DOI: 10.1038/s41522-024-00610-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
The interaction between the gut-microbiota-derived metabolites and brain has long been recognized in both health and disease. The liver, as the primary metabolic organ for nutrients in animals or humans, plays an indispensable role in signal transduction. Therefore, in recent years, Researcher have proposed the Gut-Liver-Brain Axis (GLBA) as a supplement to the Gut-Brain Axis. The GLBA plays a crucial role in numerous physiological and pathological mechanisms through a complex interplay of signaling pathways. However, gaps remain in our knowledge regarding the developmental and functional influences of the GLBA communication pathway. The gut microbial metabolites serve as communication agents between these three distant organs, functioning prominently within the GLBA. In this review, we provide a comprehensive overview of the current understanding of the GLBA, focusing on signaling molecules role in animal and human health and disease. In this review paper elucidate its mechanisms of communication, explore its implications for immune, and energy metabolism in animal and human, and highlight future research directions. Understanding the intricate communication pathways of the GLBA holds promise for creating innovative treatment approaches for a wide range of immune and metabolic conditions.
Collapse
Affiliation(s)
- Xiaoge Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
- Department of Neurosurgery, College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Manish Shukla
- Department of Neurosurgery, College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Wei Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.
| |
Collapse
|
39
|
Schneider EL, Hangasky JA, Fernández RDV, Ashley GW, Santi DV. The limitation of lipidation: Conversion of semaglutide from once-weekly to once-monthly dosing. Proc Natl Acad Sci U S A 2024; 121:e2415815121. [PMID: 39531496 PMCID: PMC11588095 DOI: 10.1073/pnas.2415815121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
The objective of this work was to develop a long-acting form of the lipidated peptide semaglutide that can be administered to humans once-monthly. Semaglutide was attached to hydrogel microspheres by a cleavable linker with an expected in vivo release half-life of about 1 mo. After a single subcutaneous dose, the pharmacokinetic parameters of released semaglutide and bodyweight loss were determined in mice, and results were used to estimate the dosing and pharmacokinetics of the released semaglutide in humans. The in vivo half-life of released semaglutide was ~36 d, and a single dose in diet-induced obese mice resulted in a lean-sparing body weight loss of 20% over 1 mo, statistically the same as semaglutide dosed twice daily. Simulations indicated the microsphere-semaglutide would permit once-monthly administration in humans; moreover, it could maintain the therapeutic minimum concentration (Cmin) of once-weekly semaglutide with only 75% of the once-weekly maximum concentration (Cmax), a feature that could reduce adverse side effects or allow higher dosing. The same approach should enable the conversion of other lipidated peptides from once-weekly to once-monthly administration.
Collapse
Affiliation(s)
| | | | | | | | - Daniel V. Santi
- ProLynx, Inc., San Francisco, CA94107
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94143
| |
Collapse
|
40
|
Garvey WT. Future Medications for Obesity and Clinical Implications. Diabetes Spectr 2024; 37:325-334. [PMID: 39649698 PMCID: PMC11623045 DOI: 10.2337/dsi24-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Semaglutide and tirzepatide have recently been approved for obesity and found to achieve ≥15% weight loss in clinical trials. These drugs have been referred to as second-generation medications because the unprecedented degree of weight loss they afford is sufficient to treat or prevent a broad array of obesity complications and related diseases. Many other medications are in development based on the actions of nutrient-regulated hormones (NRHs), including mono-, dual-, and triple-receptor agonists/antagonists for glucagon-like peptide 1, glucose-dependent insulinotropic polypeptide, amylin, peptide tyrosine-tyrosine, and glucagon. Clinical trial evidence is accumulating that these medications ameliorate multiple biomechanical, metabolic, and vascular complications of obesity. These tools enable a comprehensive complications-centric approach to care within the contextual framework of the diagnostic term adiposity-based chronic disease (ABCD). The potential to reduce patient suffering and the huge social burden of ABCD is profound. The current era of drug development based on NRHs could represent a landmark in the history of medicine provided that societies ensure access to these medications for the patients who need them.
Collapse
Affiliation(s)
- W. Timothy Garvey
- Department of Nutrition Sciences and the UAB Diabetes Research Center, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
41
|
Mendes FC, Garcia-Larsen V, Moreira A. Obesity and Asthma: Implementing a Treatable Trait Care Model. Clin Exp Allergy 2024; 54:881-894. [PMID: 38938020 DOI: 10.1111/cea.14520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/29/2024]
Abstract
Recognition of obesity as a treatable trait of asthma, impacting its development, clinical presentation and management, is gaining widespread acceptance. Obesity is a significant risk factor and disease modifier for asthma, complicating treatment. Epidemiological evidence highlights that obese asthma correlates with poorer disease control, increased severity and persistence, compromised lung function and reduced quality of life. Various mechanisms contribute to the physiological and clinical complexities observed in individuals with obesity and asthma. These encompass different immune responses, including Type IVb, where T helper 2 cells are pivotal and driven by cytokines like interleukins 4, 5, 9 and 13, and Type IVc, characterised by T helper 17 cells and Type 3 innate lymphoid cells producing interleukin 17, which recruits neutrophils. Additionally, Type V involves immune response dysregulation with significant activation of T helper 1, 2 and 17 responses. Finally, Type VI is recognised as metabolic-induced immune dysregulation associated with obesity. Body mass index (BMI) stands out as a biomarker of a treatable trait in asthma, readily identifiable and targetable, with significant implications for disease management. There exists a notable gap in treatment options for individuals with obese asthma, where asthma management guidelines lack specificity. For example, there is currently no evidence supporting the use of incretin mimetics to improve asthma outcomes in asthmatic individuals without Type 2 diabetes mellitus (T2DM). In this review, we advocate for integrating BMI into asthma care models by establishing clear target BMI goals, promoting sustainable weight loss via healthy dietary choices and physical activity and implementing regular reassessment and referral as necessary.
Collapse
Affiliation(s)
- Francisca Castro Mendes
- EPIUnit-Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional Em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
- Basic and Clinical Immunology Unit, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Vanessa Garcia-Larsen
- Program in Human Nutrition, Department of International Health, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland, USA
| | - André Moreira
- EPIUnit-Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório Para a Investigação Integrativa e Translacional Em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
- Basic and Clinical Immunology Unit, Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
- Serviço de Imunoalergologia, Centro Hospitalar Universitário São João, Porto, Portugal
| |
Collapse
|
42
|
Drucker DJ. Efficacy and Safety of GLP-1 Medicines for Type 2 Diabetes and Obesity. Diabetes Care 2024; 47:1873-1888. [PMID: 38843460 DOI: 10.2337/dci24-0003] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/14/2024] [Indexed: 10/23/2024]
Abstract
The development of glucagon-like peptide 1 receptor agonists (GLP-1RA) for type 2 diabetes and obesity was followed by data establishing the cardiorenal benefits of GLP-1RA in select patient populations. In ongoing trials investigators are interrogating the efficacy of these agents for new indications, including metabolic liver disease, peripheral artery disease, Parkinson disease, and Alzheimer disease. The success of GLP-1-based medicines has spurred the development of new molecular entities and combinations with unique pharmacokinetic and pharmacodynamic profiles, exemplified by tirzepatide, a GIP-GLP-1 receptor coagonist. Simultaneously, investigational molecules such as maritide block the GIP and activate the GLP-1 receptor, whereas retatrutide and survodutide enable simultaneous activation of the glucagon and GLP-1 receptors. Here I highlight evidence establishing the efficacy of GLP-1-based medicines, while discussing data that inform safety, focusing on muscle strength, bone density and fractures, exercise capacity, gastrointestinal motility, retained gastric contents and anesthesia, pancreatic and biliary tract disorders, and the risk of cancer. Rapid progress in development of highly efficacious GLP-1 medicines, and anticipated differentiation of newer agents in subsets of metabolic disorders, will provide greater opportunities for use of personalized medicine approaches to improve the health of people living with cardiometabolic disorders.
Collapse
Affiliation(s)
- Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| |
Collapse
|
43
|
Riehl-Tonn VJ, Medak KD, Rampersad C, MacPhee A, Harrison TG. GLP-1 Agonism for Kidney Transplant Recipients: A Narrative Review of Current Evidence and Future Directions Across the Research Spectrum. Can J Kidney Health Dis 2024; 11:20543581241290317. [PMID: 39492845 PMCID: PMC11528610 DOI: 10.1177/20543581241290317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/25/2024] [Indexed: 11/05/2024] Open
Abstract
Purpose of Review Diabetes is the most common cause of kidney disease in individuals that receive a kidney transplant, and those without pre-existing diabetes are at greater risk of developing diabetes following kidney transplant. A class of diabetes treatment medications called glucagon-like peptide-1 receptor agonists (GLP-1RA) has seen recent widespread use for people with diabetes or obesity, with efficacy for improved glycemic control, weight loss, and reduced risk of cardiovascular events. Given these benefits, and indications for use that often co-occur in kidney transplant recipients, use of GLP-1RAs warrants consideration in this population. Therefore, we sought to review the current literature to better understand the mechanisms of action, clinical application, and person-centred considerations of GLP-1RAs in kidney transplant recipients. Sources of Information Original articles were identified between December 2023 and July 2024 from electronic databases including the Ovid MEDLINE database, PubMed, and Google Scholar using terms "kidney transplant," "GLP-1," "glucagon-like peptide-1 receptor agonist," and "diabetes." Methods A comprehensive review of the literature was conducted to explore the relationship between GLP-1RAs and kidney transplant recipients. We reviewed the current state of evidence across the research disciplines of basic or fundamental science, clinical and health services research, and person-centred equity science, and highlighted important knowledge gaps that offer opportunities for future research. Key Findings Numerous clinical studies have demonstrated the benefit of GLP-1RAs in people with and without diabetic kidney disease, including decreased risk of cardiovascular events. However, there is a paucity of high-quality randomized controlled trials and observational studies analyzing use of GLP-1RAs in kidney transplant recipients. Evidence of benefit in this population is therefore limited to small studies or inferred from research conducted in nontransplant populations. Growing evidence from preclinical and clinical studies may elucidate renoprotective mechanisms of GLP-1RAs and remove barriers to application of these drugs in the transplant recipient population. Individuals who are female, non-white, have lower socioeconomic status, and live in rural communities are at greater risk of diabetes and have lower uptake of GLP-1RAs. There is a need for clinical trials across diverse kidney transplant populations to estimate the efficacy of GLP-1RAs on important health outcomes. Limitations The search strategy for this narrative review may not have been sensitive to identify all relevant articles. Our search was limited to English language articles.
Collapse
Affiliation(s)
- Victoria J. Riehl-Tonn
- Department of Medicine, University of Calgary, AB, Canada
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, AB, Canada
| | - Kyle D. Medak
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Christie Rampersad
- Ajmera Transplant Centre, Toronto General Hospital, University Health Network, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, ON, Canada
| | - Anne MacPhee
- Canadians Seeking Solutions and Innovations to Overcome Chronic Kidney Disease (Can-SOLVE CKD), Vancouver, BC, Canada
| | - Tyrone G. Harrison
- Department of Medicine, University of Calgary, AB, Canada
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, AB, Canada
- Department of Community Health Sciences, University of Calgary, AB, Canada
- O’Brien Institute for Public Health, Cumming School of Medicine, University of Calgary, AB, Canada
| |
Collapse
|
44
|
Gill D, Dib MJ, Cronjé HT, Karhunen V, Woolf B, Gagnon E, Daghlas I, Nyberg M, Drakeman D, Burgess S. Common pitfalls in drug target Mendelian randomization and how to avoid them. BMC Med 2024; 22:473. [PMID: 39407214 PMCID: PMC11481744 DOI: 10.1186/s12916-024-03700-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Drug target Mendelian randomization describes the use of genetic variants as instrumental variables for studying the effects of pharmacological agents. The paradigm can be used to inform on all aspects of drug development and has become increasingly popular over the last decade, particularly given the time- and cost-efficiency with which it can be performed even before commencing clinical studies. MAIN BODY In this review, we describe the recent emergence of drug target Mendelian randomization, its common pitfalls, how best to address them, as well as potential future directions. Throughout, we offer advice based on our experiences on how to approach these types of studies, which we hope will be useful for both practitioners and those translating the findings from such work. CONCLUSIONS Drug target Mendelian randomization is nuanced and requires a combination of biological, statistical, genetic, epidemiological, clinical, and pharmaceutical expertise to be utilized to its full potential. Unfortunately, these skillsets are relatively infrequently combined in any given study.
Collapse
Affiliation(s)
- Dipender Gill
- Sequoia Genetics, London, UK.
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, 90 Wood Lane, London, W12 0BZ, UK.
| | - Marie-Joe Dib
- Cardiovascular Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Héléne T Cronjé
- Sequoia Genetics, London, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Ville Karhunen
- Sequoia Genetics, London, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Benjamin Woolf
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- School of Psychological Science, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Eloi Gagnon
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Québec, Canada
| | - Iyas Daghlas
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Michael Nyberg
- Cardiovascular Biology, Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | - Donald Drakeman
- University of Cambridge Centre for Health Leadership & Enterprise, Judge Business School, Trumpington Street, Cambridge, UK
- Advent Venture Partners, London, UK
| | - Stephen Burgess
- Sequoia Genetics, London, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
45
|
Kukova L, Munir KM, Sayeed A, Davis SN. Assessing the therapeutic and toxicological profile of novel GLP-1 receptor agonists for type 2 diabetes. Expert Opin Drug Metab Toxicol 2024; 20:939-952. [PMID: 39268978 DOI: 10.1080/17425255.2024.2401589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
INTRODUCTION GLP-1 receptor agonists provide multiple benefits for patients with type 2 diabetes. Nonetheless, there are also several significant adverse effects associated with these agents. A thorough understanding of both therapeutic and toxicological profiles of GLP-1 receptor agonists is crucial for appropriate utilization of this medication class. A literature search of PubMed and ClinicalTrials.gov was carried out to inform discussion on the topic. AREAS COVERED This review article discusses the key advantages and disadvantages derived from the use of GLP-1 receptor agonists in the treatment of type 2 diabetes. Landmark trials which helped characterize the cardiovascular and renal benefits of GLP-1 receptor agonists are highlighted. We also discuss key studies still in progress and new formulations under investigation. EXPERT OPINION GLP-1 receptor agonists provide glycemic and complication-risk reduction benefits for individuals with type 2 diabetes. Current data suggests there is a lot of potential for further applications, even outside of type 2 diabetes management. It would be of particular interest to see the range of benefits conferred from GLP-1 receptor agonists in individuals without type 2 diabetes. Broader application of these medications could be expected given the ongoing development of new oral formulations and combination agents.
Collapse
Affiliation(s)
- Lidiya Kukova
- Internal Medicine Resident, Department of Internal Medicine, University of Maryland Medical Center, Baltimore, MD, USA
| | - Kashif M Munir
- Professor of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland Medical Center, Baltimore, MD, USA
| | - Ahmed Sayeed
- Medical Student, American University of Antigua College of Medicine, Coolidge, Anitgua and Barbuda
| | - Stephen N Davis
- Chair, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
46
|
Khezri H, Mostafavi M, Dabirmanesh B, Khajeh K. Peptibodies: Bridging the gap between peptides and antibodies. Int J Biol Macromol 2024; 278:134718. [PMID: 39142490 DOI: 10.1016/j.ijbiomac.2024.134718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Peptides are a very critical class of pharmaceutical compounds that can control several signaling pathways and thereby affect many physiological and biochemical processes. Previous research suggests that both peptides and antibodies may serve as potent tools for research, diagnostics, vaccination, and therapeutics across diverse domains. The distinct attributes of peptides, like their profound tissue penetration, efficient cellular internalization, reduced immunogenicity, and adaptability to chemical modification, underscore their significance in biomedical applications. However, they also possess drawbacks such as lower affinity, poor absorption, low stability to proteolytic digestion, and rapid clearance. The advent of peptibodies is a significant advance that improves the limitations of both peptides and antibodies. Peptibodies, or Peptide-Fc fusions, represent a promising therapeutic modality comprising biologically active peptides fused to an Fc domain. The stability and efficacy of the peptide are enhanced by this fusion strategy, which overcomes some of the inherent limitations. Many peptibodies have been developed to treat conditions like cancer, diabetes, and lupus. Romiplostim and Dulaglutide are the only ones approved by the EMA and FDA, respectively. Given the growing significance of peptibodies in the pharmaceutical landscape, this investigation aims to explain key aspects encompassing the intrinsic properties of peptides, the intricacies of peptibody production, and their potential therapeutic applications.
Collapse
Affiliation(s)
- Hamidhossein Khezri
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdiyeh Mostafavi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
47
|
Holst JJ. GLP-1 physiology in obesity and development of incretin-based drugs for chronic weight management. Nat Metab 2024; 6:1866-1885. [PMID: 39160334 DOI: 10.1038/s42255-024-01113-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024]
Abstract
The introduction of the highly potent incretin receptor agonists semaglutide and tirzepatide has marked a new era in the treatment of type 2 diabetes and obesity. With normalisation of glycated haemoglobin levels and weight losses around 15-25%, therapeutic goals that were previously unrealistic are now within reach, and clinical trials have documented that these effects are associated with reduced risk of cardiovascular events and premature mortality. Here, I review this remarkable development from the earliest observations of glucose lowering and modest weight losses with native glucagon-like peptide (GLP)-1 and short acting compounds, to the recent development of highly active formulations and new molecules. I will classify these agents as GLP-1-based therapies in the understanding that these compounds or combinations may have actions on other receptors as well. The physiology of GLP-1 is discussed as well as its mechanisms of actions in obesity, in particular, the role of sensory afferents and GLP-1 receptors in the brain. I provide details regarding the development of GLP-1 receptor agonists for anti-obesity therapy and discuss the possible mechanism behind their beneficial effects on adverse cardiovascular events. Finally, I highlight new pharmacological developments, including oral agents, and discuss important questions regarding maintenance therapy.
Collapse
Affiliation(s)
- Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences. Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
48
|
Manchanda Y, Tomas A. New insights into the regulation of GIPR signalling. Nat Rev Endocrinol 2024; 20:571-572. [PMID: 39174673 DOI: 10.1038/s41574-024-01027-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Affiliation(s)
- Yusman Manchanda
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
49
|
Jensen MH, Sanni SJ, Riber D, Holst JJ, Rosenkilde MM, Sparre-Ulrich AH. AT-7687, a novel GIPR peptide antagonist, combined with a GLP-1 agonist, leads to enhanced weight loss and metabolic improvements in cynomolgus monkeys. Mol Metab 2024; 88:102006. [PMID: 39128651 PMCID: PMC11382121 DOI: 10.1016/j.molmet.2024.102006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024] Open
Abstract
OBJECTIVES Obesity represents a global health crisis with significant patient burdens and healthcare costs. Despite the advances with glucagon-like peptide-1 (GLP-1) receptor agonists in treating obesity, unmet needs remain. This study characterizes a novel glucose-dependent insulinotropic polypeptide receptor (GIPR) peptide antagonist, AT-7687, evaluating its potential to enhance obesity treatment. METHODS We assessed the in vitro potency and pharmacokinetics of AT-7687, alongside its therapeutic effects when administered subcutaneously (SC) alone and in combination with liraglutide to high-fat-diet-fed obese non-human primates (NHP). The study spanned a 42-day treatment period and a 15-day washout period. RESULTS AT-7687 demonstrated a subnanomolar cAMP antagonistic potency (pKB of 9.5) in HEK-293 cells and a 27.4 h half-life in NHPs. It effectively maintained weight stability in obese monkeys, whereas placebo recipients had an 8.6% weight increase by day 42 (P = 0.01). Monotherapy with liraglutide resulted in a 12.4% weight reduction compared to placebo (P = 0.03) and combining AT-7687 with liraglutide led to a 16.3% weight reduction (P = 0.0002). The combination therapy significantly improved metabolic markers, reducing insulin levels by 52% (P = 0.008), glucose by 30% (P = 0.02), triglycerides by 39% (P = 0.05), total cholesterol by 29% (P = 0.03), and LDL cholesterol by 48% (P = 0.003) compared to placebo. AT-7687 treatment was well tolerated and not associated with any side effects. CONCLUSIONS This study underscores the potential of AT-7687 as a promising addition to current obesity treatments.
Collapse
Affiliation(s)
- Mette H Jensen
- Antag Therapeutics Aps, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| | - Samra J Sanni
- Antag Therapeutics Aps, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| | - Ditte Riber
- Antag Therapeutics Aps, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Alle 14, 2200 Copenhagen N, Denmark
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Alle 14, 2200 Copenhagen N, Denmark
| | | |
Collapse
|
50
|
Prentza V, Pavlidis G, Ikonomidis I, Pililis S, Lampsas S, Kountouri A, Pliouta L, Korakas E, Thymis J, Palaiodimou L, Tsegka A, Markakis K, Halvatsiotis P, Tsivgoulis G, Lambadiari V. Antidiabetic Treatment and Prevention of Ischemic Stroke: A Systematic Review. J Clin Med 2024; 13:5786. [PMID: 39407846 PMCID: PMC11476433 DOI: 10.3390/jcm13195786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Diabetes mellitus (DM) is a prevalent disease in the general population and also a well-established risk factor for the development of ischemic stroke. Patients who have been diagnosed with diabetes have a 20% higher risk for developing ischemic stroke in comparison to non-diabetic individuals. The aim of the current systematic review is to provide the latest evidence regarding the association between antidiabetic treatment and the prevention of ischemic stroke. Methods: A comprehensive search in scientific literature databases PUBMED, COCHRANE, and SCOPUS was conducted. The studies that were deemed as eligible for this review were those that examined the clinical benefits of therapeutic strategies in terms of preventing ischemic strokes. Results: A total of 32 studies met the established selection criteria. The included studies showed that pioglitazone treatment significantly reduced the risk for recurrent stroke in patients with DM. Furthermore, in the context of primary prevention, the improvement in glycemic control after treatment with the glucagon-like peptide-1 receptor agonists (GLP-1RA) semaglutide and dulaglutide was associated with a reduction in the risk of ischemic stroke in diabetic subjects. Metformin monotherapy may reduce stroke risk, while dipeptidyl peptidase 4 inhibitors, sodium-glucose co-transporter 2 inhibitors, and insulin do not seem to affect the incidence of stroke. Conclusions: The findings of the present systematic review suggest that pioglitazone and GLP-1RA may decrease the risk of stroke. Further studies are needed to provide additional data regarding the preventive effect of novel antidiabetic drugs, such as dual glucose-dependent insulinotropic polypeptide/GLP-1RA agents, on stroke.
Collapse
Affiliation(s)
- Vasiliki Prentza
- 2nd Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (V.P.); (I.I.); (S.L.); (J.T.)
| | - George Pavlidis
- 2nd Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (V.P.); (I.I.); (S.L.); (J.T.)
| | - Ignatios Ikonomidis
- 2nd Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (V.P.); (I.I.); (S.L.); (J.T.)
| | - Sotirios Pililis
- Research Unit and Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (L.P.); (E.K.); (A.T.); (K.M.); (P.H.); (V.L.)
| | - Stamatios Lampsas
- 2nd Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (V.P.); (I.I.); (S.L.); (J.T.)
| | - Aikaterini Kountouri
- Research Unit and Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (L.P.); (E.K.); (A.T.); (K.M.); (P.H.); (V.L.)
| | - Loukia Pliouta
- Research Unit and Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (L.P.); (E.K.); (A.T.); (K.M.); (P.H.); (V.L.)
| | - Emmanouil Korakas
- Research Unit and Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (L.P.); (E.K.); (A.T.); (K.M.); (P.H.); (V.L.)
| | - John Thymis
- 2nd Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (V.P.); (I.I.); (S.L.); (J.T.)
| | - Lina Palaiodimou
- 2nd Department of Neurology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece (G.T.)
| | - Aikaterini Tsegka
- Research Unit and Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (L.P.); (E.K.); (A.T.); (K.M.); (P.H.); (V.L.)
| | - Konstantinos Markakis
- Research Unit and Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (L.P.); (E.K.); (A.T.); (K.M.); (P.H.); (V.L.)
| | - Panagiotis Halvatsiotis
- Research Unit and Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (L.P.); (E.K.); (A.T.); (K.M.); (P.H.); (V.L.)
| | - Georgios Tsivgoulis
- 2nd Department of Neurology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece (G.T.)
| | - Vaia Lambadiari
- Research Unit and Diabetes Center, 2nd Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (L.P.); (E.K.); (A.T.); (K.M.); (P.H.); (V.L.)
| |
Collapse
|